Next Article in Journal
RB1-Negative Retinal Organoids Display Proliferation of Cone Photoreceptors and Loss of Retinal Differentiation
Previous Article in Journal
7T HR FID-MRSI Compared to Amino Acid PET: Glutamine and Glycine as Promising Biomarkers in Brain Tumors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Antitumor Properties of Curcumin in Breast Cancer Based on Preclinical Studies: A Systematic Review

by
Kênia Alves Barcelos
1,2,*,
Carolina Rodrigues Mendonça
3,
Matias Noll
3,4,5,
Ana Flávia Botelho
1,
Cristiane Raquel Dias Francischini
1 and
Marco Augusto Machado Silva
1,*
1
Postgraduate Program of Animal Science, Escola de Veterinária e Zootecnia, Universidade Federal de Goiás, Goiás 74690-900, Brazil
2
Faculty of Medicine, Universidade de Rio Verde—UniRV, Rio de Janeiro 75901-970, Brazil
3
Graduate Program in Health Sciences, Escola de Medicina, Universidade Federal de Goiás, Goiás 74605-050, Brazil
4
Instituto Federal Goiano (IF Goiano), Goiás 74270-040, Brazil
5
Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, 5230 Odense, Denmark
*
Authors to whom correspondence should be addressed.
Cancers 2022, 14(9), 2165; https://doi.org/10.3390/cancers14092165
Submission received: 2 March 2022 / Revised: 14 April 2022 / Accepted: 18 April 2022 / Published: 26 April 2022

Abstract

:

Simple Summary

Natural formulations and phytotherapies have shown promising antitumor activities. This review assesses the antitumor effects of curcumin on breast cancer. In particular, we discuss the effects of curcumin on the proliferation, viability, and apoptosis of breast cancer cell lineages and tumor volume. Studies have shown that curcumin administered at different concentrations inhibited proliferation, decreased viability, and induced apoptosis in human and animal breast cancer cells. Nanoparticle formulations of curcumin administered orally, via implant, or intraperitoneally reduced the tumor volume of human and murine mammary cells in vivo. Moreover, curcumin nanoformulations facilitate tumor growth inhibition in animal models of breast cancer. Randomized clinical trials are warranted to assess the efficacy and safety of curcumin formulations for clinical use.

Abstract

Breast cancer is one of the most common neoplasms among women. Anticancer strategies using natural formulations and phytotherapies are promising antitumor treatment alternatives. This review assesses the antitumor effects of curcumin on breast cancer reported in preclinical in vitro and in vivo animal models. We used five databases to search for preclinical studies published up to May 2021. The assessments included the effects of curcumin on the proliferation, viability, and apoptosis of breast cancer cell lineages and on tumor volume. In total, 60 articles met the inclusion criteria. Curcumin administered at different concentrations and via different routes of administration inhibited proliferation, decreased viability, and induced apoptosis in human and animal breast cancer cells. Nanoparticle formulations of curcumin administered orally, via implant, and intraperitoneally reduced the tumor volume of human and murine mammary cells in vivo. Moreover, curcumin nanoformulations exert positive effects on tumor growth inhibition in animal models of breast cancer. Further randomized clinical trials are warranted to assess the efficacy and safety of curcumin formulations for clinical use.

Graphical Abstract

1. Introduction

Breast cancer is one of the most common neoplasms in women and an important public health problem worldwide [1]. Breast cancer has surpassed lung neoplasm as the most frequently diagnosed cancer, with approximately 2.3 million new cases reported (11.7%) in 2020, according to the Global Cancer Observatory [2].
Conventional treatment for breast cancer includes surgical resection, radiotherapy, and chemotherapy [3]. In addition, promising alternative approaches, such as targeted therapy, immunotherapy, and hormone therapy, are currently under investigation [4,5]. These therapies vary in their mechanisms of action. The appropriate treatment regime is determined based on the type of tumor, disease stage, and clinical condition of patients [4,5].
Although chemotherapy remains the gold standard for treating several types of cancer, severe adverse reactions and tumor resistance to treatment and hormone therapy are considered negative aspects of paramount importance [3,6]. Therefore, alternative anticancer therapeutic strategies, such as the use of low-toxicity natural subproducts and extracts, are promising modalities [6,7].
Previous studies have reported that curcumin, a turmeric-derived phytochemical, exhibits beneficial biological activities, including antibacterial, antiviral, anticancer, anti-inflammatory, and antioxidant properties, and was found to exert preventive and therapeutic effects in various cancers, including breast cancer [7,8,9,10,11,12]. However, the therapeutic applicability of curcumin remains limited owing to its low water solubility and bioavailability [7,13]. Only two systematic reviews on the effects of curcumin on breast cancer have been reported to date [13,14]. Gianfredi et al. [14] investigated the bioactive effects of a curcumin-containing diet on human breast cancer cell lines [14]. Meanwhile, Ombredane et al. [13] reported the in vivo efficacy and toxicity of curcumin nanoparticles (CUR-NPs) as a treatment strategy against breast cancer. Therefore, there remain gaps in the literature regarding the effects of curcumin on tumors. In this systematic review, we collated data from preclinical in vitro and in vivo studies conducted on animal models to investigate the effects of curcumin on the proliferation, viability, and apoptosis of breast cancer cells and tumor volume, focusing on dose and administration route. We systematically reviewed the antitumor effects of curcumin on breast cancer previously reported. To our knowledge, this is the first systematic review of in vitro preclinical studies on the effect of curcumin on breast cancer cell lineages and animal models of breast cancer.

2. Materials and Methods

2.1. Protocol and Registration

The study design was based on the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses [15] and the Systematic Review Center for Laboratory Animal Experimentation (SYRCLE) [16]. A protocol was published in the International Prospective Register of Systematic Reviews: Review of animal data from experimental studies databases (CRD42021256605). We included data from preclinical in vitro and in vivo studies conducted using animal models.

2.2. Search Strategy and Eligibility Criteria

The PubMed, Embase, Scopus, Web of Science, and SciELO databases were used for data retrieval. The research period was limited to 23 May 2021. Google Scholar and the reference lists of primary studies were consulted to search for additional studies. The following uniterms were used: “Curcumin”; “Curcuma longa”; “Turmeric”; “Natural yellow 3”; “Turmeric yellow”; “Indian saffron”; “Kacha haldi”; “Curcumin Nanoparticles”; “Breast Cancer”; “Breast Neoplasms”; “Triple Negative”; “Breast Neoplasms”; “Breast Tumor”; “Inflammatory Breast Neoplasms”; “Carcinoma, Ductal, Breast”; “Carcinoma Lobular”; “Her-2 Positive”; “Breast Cancer”; “In Vitro”; “Mouse”; “Animal”. The search strategy adopted for each database is listed in (Table S1).
The participants, intervention, comparator, and outcomes (PICO) framework was used to determine the eligibility criteria for the systematic review of preclinical animal studies, as follows:
  • Patient: laboratory animals with induced breast cancer (all species).
  • Intervention(s): curcumin
  • Comparator(s): control group or comparison with no treatment, treatment with other drugs, and/or traditional radiotherapy or chemotherapy regimens.
  • Outcomes: antitumor activity (reduction in tumor volume and dimensions) in in vitro studies.
The inclusion criteria were as follows: (1) in vitro and animal experimental model investigations on the effects of curcumin on human and animal breast cancer cells of different lineages, (2) peer-reviewed original research articles, (3) no language restrictions, and (4) no publication year restriction. The exclusion criteria were as follows: (1) doctoral and master’s theses, (2) case studies, (3) editorials, (4) letters to editor, (5) duplicate studies found in more than one database and in silico studies, (6) epidemiological studies, (7) clinical assays and articles that requested permission from authors without response, (8) studies irrelevant to the antitumor effect of curcumin on breast cancer, (9) trials performed in non-oncological clinical conditions, (10) studies involving a sole treatment protocol based on the association between curcumin and other treatment modalities, and (11) trials involving immunodeficient animal models.

Definitions

Cell proliferation: increase in cell count owing to cell division [17]. Cell proliferation was strictly controlled without any alterations. In contrast, neoplastic cells exhibited massive and uncontrolled proliferation [18].
Cell viability: quantification of viable cells for estimating cytotoxicity [19] and investigating cell activity and integrity [19,20].
Apoptosis: programmed cell death under physiological and pathological conditions [21,22]. In cancer, disparity between cell replication and death causes malignancy [22].

2.3. Review Process

Two authors (K.A.B. and C.R.M.) performed a peer review of the titles and abstracts of the articles using Rayyan software. The selected articles were assessed by the authors and critically evaluated based on the known antitumor effects of curcumin on breast cancer. Next, the selected articles were assessed and the inclusion/exclusion criteria were applied. Doubts and disagreements regarding article selection were discussed with the research team. If some published studies were associated with the same project or were retrieved from the same database, the most complete study was selected [23,24].

2.4. Training of Reviewers

The authors participating in eligibility assessments completely understood each step of the review process, primarily the inclusion/exclusion criteria, and practiced eligibility assessments on 50 test abstracts prior to coding articles. The authors also used risk-of-bias instruments and performed quality assessments and data extraction on five articles that were not included in the review [23].

2.5. Evidence Synthesis

The following data were extracted: authorship, year of publication, country, cell lineage, concentration, exposure time, animal experimental model, follow-up, sample, dosing, route of administration, and main outcomes. The outcomes included antitumor activity, including cell proliferation, viability, apoptosis, and/or cessation of the cell cycle in in vitro studies and changes in tumor volume and magnitude in animal models.
The Grading of Recommendations Assessment, Development, and Evaluation (GRADE) tool, adapted for in vitro study designs [25], was used to assess quality, since methods specifically for this purpose are lacking. In vitro trials were ranked as “high,” “moderate,” and “low” in terms of quality [25] based on the analysis of each study.
The SYRCLE RoB Toll tool was used to assess the quality of animal model studies [26]. Selection, performance, detection, attrition, reporting, and other biases were investigated.
There was substantial heterogeneity among the studies, which was detrimental to meta-analysis. Therefore, narrative synthesis was performed without statistical or sensitivity analysis, assessing publication bias using the funnel plot and Egger’s and Begg’s tests.

3. Results

The bibliographic survey yielded 1288 articles. After titles and abstracts from the records were screened, 104 potentially eligible articles were identified and selected for complete reading. Following a review of all texts, 44 articles were excluded. Details of the search strategies are provided in (Table S1). The reasons for exclusion are provided in (Table S2). The flowchart of the study is shown in Figure 1.

3.1. General Characteristics of the Studies

Sixty studies on the effect of curcumin on breast cancer [3,7,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84] were included in this investigation, with 23 in vitro trials [27,28,31,33,34,35,36,37,44,45,48,51,52,54,59,64,65,70,73,74,76,80,84], 20 studies on animal models [29,30,32,40,41,46,57,60,62,66,68,69,71,72,75,77,79,82,83], and 17 studies with both in vitro and in vivo experimental designs [3,7,39,42,43,47,49,50,53,55,56,58,61,63,67,78,81]. The oldest and most recent articles were published in 1997 [64] and 2021 [39,81], respectively. The general characteristics of the selected articles are presented in Table 1 and Table 2.

3.2. Summary of the Results

3.2.1. In Vitro Studies

Forty studies were conducted using in vitro design and assessment (Table 1) [3,7,27,28,31,33,34,35,36,37,39,42,43,44,45,47,48,49,50,51,52,53,54,55,56,58,59,61,63,64,65,67,70,73,74,76,79,80,81,84].
The human breast cancer cell lineages used in the studies were as follows: MCF-7 [27,31,34,36,42,44,48,49,51,56,64,65,70,76,78,79], MDA-MB-435 [45,63], T47D [35,44], MCF-7/LCC2 [48], LCC9 [48], MDA-MB-468 [50,63], and BT-474 [63].
Moreover, studies conducted using the triple-negative breast cancer cell line MDA-MB-231 [3,28,31,33,36,37,39,47,52,53,54,56,59,61,63,67,73,74] and human breast cancer cell lineage expressing the Her2 SK-BR-3 gene [63,80] were also assessed. In animal models, murine mammary carcinoma 4T1 [43,53,58,81] and H-2” (TUBO) [63] cell lineages were investigated.

3.2.2. In Vitro Cell Proliferation

The in vitro proliferation of breast cancer cell lineages was assessed using a quantitative image assessment technique [34], Transwell assay [42], colony formation assay [48,50], sulforhodamine B, colorimetric analysis [56], method (NR) for determining inhibition of cell growth [59], thymidine incorporation assay [3H], flow cytometry tests [64], and MTT assay, as described in most studies (Table 1).
The effect of curcumin on cell proliferation was investigated only in human cell lines. Curcumin administered at concentrations of 1, 3, 10, 20, 30, and 50 μg/mL for 24 h inhibited the proliferation of MCF-7 cells, with growth recurrence in the subsequent 72 h [27,34,42,44,56,64,76]. Optimal inhibition was achieved upon treatment with a single dose of 25 μM curcumin for 24 h [34]. A substantial reduction in growth was observed in malignant MCF-7 cell lines, with 37%, 54%, and 73% reduction upon treatment with 20, 50, and 100 μM curcumin, respectively [84].
Proliferation in MDA-MB-435 cell lineages was inhibited following treatment with 0, 10, 25, 50, and 75 μM curcumin [45,63]. The formation of colonies from MCF-7/LCC2 cells was inhibited following treatment with 30 μM curcumin [48]. The number of colonies in MDA-MB-468 cell cultures reduced over two weeks upon treatment with 5 µM curcumin [50]. Likewise, the proliferation of BT-474 cell cultures was inhibited upon treatment with 10 μg/mL curcumin [56]. In studies on triple-negative MDA-MB-231 cell lineages, cell proliferation was inhibited upon treatment with 0, 1, 25, 2,5, 5, 10, 15, 20, 30, and 50 μM curcumin for 24 and 48 h [7,28,33,59,73]. Furthermore, MSN-curcumin nanoparticles exhibited anticancer properties at 20 μg/mL [39].
Table 1. Characteristics of the in vitro studies included in the systematic review on curcumin and breast cancer.
Table 1. Characteristics of the in vitro studies included in the systematic review on curcumin and breast cancer.
Author/Year/CountryType of Cell/ModelInterventionOutcomesConflict of Interest
Antitumor Activity
Concentration (Component)Treatment DurationCell Proliferation In VitroCell ViabilityApoptosis and/or Cell Cycle Interruption
Abbaspour and Afshar, 2018 [27]
Iran
MCF-7
Human
Curcumin at
10, 20 and 30 μg/mL
24, 48, and 72 hMTT assay
↓ cell proliferation owing to downregulation of ODC1 and ADA gene expression.
MTT assay
↓ viability of cells in a time- and dose-dependent manner.
Not reportedNone
Abuelba et al., 2015 [28]
Romania
MDA-MB-231
Human
Curcumin at
15–19 μM
24, 48, and 72 hMTT assay
↓ cell proliferation upon treatment with 15 μM curcumin.
MTT assay
↓ cell viability by up to 25% upon treatment with 15 μM curcumin.
MTT assay
Pro-apoptotic effects on MDA-MB-231 cells cultured in a single layer, without photoactivation.
None
Bimonte et al., 2015 [7]
Italy
MDA.MB231
Human
Curcumin at
10 and 50 μM
48 hMTT assay
Inhibition of breast cancer cell migration in 48 h.
↓ cell proliferation (p < 0.05).
Not reportedFlow cytometry
Curcumin (10 μM) ↑ apoptosis (p < 0.0001).
None
Calaf et al., 2018 [31]
Chile
MCF7
MDA-MB-231
Human
Curcumin at
30 µM
48 hNot reportedNot reportedFlow cytometry
Apoptosis
MDA-MB-231: 14.2%
MCF7: 4.6%
None
Chiu and Su, 2009 [33]
China
MDA-MB-231
Human
Curcumin at
10, 20, and 30 μg/mL
48 hMTT Assay
↓ proliferation of MDA-MB-231 cells via p21 expression.
Not reportedFlow cytometry
Curcumin induced apoptosis via positive regulation of the Bax:Bcl-2 ratio.
None
Choudhuri et al., 2002 [34]
India
MCF-7
Human
Curcumin at
10 and 25 μM
24 hQuantitative image analysis
Cessation of cell growth followed by significant cell death.
Optimal inhibition was obtained upon treatment with 25 μM curcumin.
Not reportedQuantitative image analysis techniques
Curcumin induced apoptosis.
None
Coker-Gurkan et al., 2019 [35]
Turkey
T47D
Human
Curcumin at 30 µM24 and 48 hNot reportedMTT assay
↓ cell viability by 48% and 60% upon treatment with 20 µM curcumin (p < 0.0024).
Double staining with Annexin-V/PI
Curcumin induced apoptosis in 10.9% and 5.2% of the cell populations.
None
Coker-Gurkan et al., 2018 [36]
Turkey
MCF-7
MDA-MB-231
Human
Curcumin at 30 µM24 and 48 hNot reportedMTT assay
↓ cell viability
MCF-7 cells by 49% and of
MDA-MB-453 cells by 48% upon treatment for 24 h with 20 µM curcumin
MTT assay
Curcumin induced apoptotic cell death.
None
Fan et al., 2016 [37]
China
MDA-MB-231
Human
Curcumin at 50 μg/mL24 hNot reportedMTT assay
↓ cell viability (% NR) (P:NR)
MTT assay
Curcumin induced apoptosis.
None
Ghosh et al., 2021 [39]
India
MDA-MB 231
Human
Curcumin at 50 μg/mL
Nanostructured platform
Nanoparticles, MSN-Curcumin (MSN-C), and MSN-Hyaluronic acid-Curcumin (MSN-HA-C)
48 hMTT Assay
MSN-HA-C blocked cell proliferation, in contrast to free curcumin. The treatment agent exhibited anticancer properties at 20 μg/mL.
Not reportedMTT assay
Cell death
MSN-HA-C: 58%
MSN-C: 34%
(with equivalent dose of 12 μg/mL curcumin).
MDA-MB-231 cycle arrest
↓ G1-phase cells:
32.5%
Control:
54.6%
G2/M phase cells: 37.8%
Controls: 11.4%.
None
Hashemzehi et al., 2018 [42]
Iran
MCF-7
Human
Curcumin at
1 mM
Nanostructured platform
Nano-curcumin—phytosomalcurcumin
24 hTranswell assay
↓ cell invasion
MTT assay
↓ cell growth in a dose-dependent manner.
Not reportedNot reportedNone
He et al., 2019 [43]
China
4T1
Mouse
Curcumin at
50 μg/mL
Nanostructured platform
Polymeric micellar NPs [amphiphilic diblock copolymer—mPEG-b-PLG (Se) -TP]
48 hNot reportedMTT assay
↓ of cell viability upon treatment with CUR-NP and
Free CUR:
15%
Not reportedNone
Hu et al., 2018 [44]
China
T47D, MCF7
Human
Curcumin at
10 or 30 µM
72 hMTT assay
↓ cell proliferation
Not reportedFlow cytometry
Apoptosis
T47D cells: 13.87% and 30.09%.
MCF7 cells: 15.14% and 35.04%.
None
Hua et al., 2010 [45]
China
MDA-MB-435
Human
Curcumin at 10, 25, 50, and 75 μM12, 24, or 48 h.MTT assay
↓ cell proliferation, inducing arrest in the G1 phase.
Not reportedNot reportedNR
Ji et al., 2020 [47]
China
MDA-MB-231
Human
Curcumin at
50 μg/mL
24 hNot reportedNot reportedFlow cytometry
Apoptosis
HA@CUR-NCs
80%.
None
Jiang et al., 2013 [48]
China
MCF-7/LCC2 and LCC9
Human
Curcumin at
10 and 30 μM
24, 48, 72, and 96 hColony formation assay
↓ colony formation Complete suppression of colony formation upon treatment with 30 μM curcumin.
Not reportedAnnexin-V/PI staining and flow cytometry
30 μM curcumin caused a significant increase (28.72% in MCF-7 cells, 31.36% in MCF-7/LCC2 cells, and 34.70% in MCF-7/LCC9 cells) in the percentage of late apoptotic cells.
None
Jin et al., 2017 [49]
China and USA
MCF-7
Human
Curcumin at
10 µg/mL
Nanostructured platform
CUR-NP;
GE11-CUR-NP; Free CUR
24 hNot reportedNanostructured platform
CUR-NP,
GE11-CUR-NP, and Free CUR
Flow cytometry
Apoptosis
CUR-NP: 14.9%;
GE11-CUR-NP: 18.9%; Free CUR 11.0%.
None
Jung et al., 2018 [50]
South Korea
MDA-MB-468
Human
Curcumin at
5 and 10 μM
72 and 96 hColony formation assay
↓ number of colonies over 2 weeks to 36.9 ± 7.7%
upon treatment with 5 µM curcumin.
Unclear method
↓ significantly decreased cell viability (41.5 ± 2.8% of basal level) upon treatment with
10 µM curcumin
Not reportedNone
Kim et al., 2012 [51]
Coreia do Sul
MCF-7
Human
Curcumin at
1, 5, 10, 30, and 50 μM
24 hNot reportedMTT assay
Curcumin exerted no effect on the viability of MCF-7 cells
Not reportedNone
Kumari et al., 2017 [52]
India
MDA-MB-231
Human
Curcumin at
50 and 100 μg/mL
Nanostructured platform
free CUR and CUR-mPEG-PLA-Ch micelles
24 hNot reportedMTT assay
CUR: 55.26 ± 3.7%
Free CUR: 66.84 ± 2.4%
(p = 0.079)
Not reportedNone
Kumari et al., 2020 [53]
India
MDA-MB-231
Human
4T1
Mouse
Curcumin at
50 μg/mL
Nanostructured platform
CUR treatment (Free CUR group—24 μg/mL) and CUR-HSA-DOPE NPs treatment (CUR-HSA-DOPE group)
6 and 24 hNot reportedMTT Assay
MDA-MB-231
Cur-HSA-DOPE NPs
24.34 ± 6.1% and 33.99 ± 4.5%
free CUR
34.87 ± 4.9% and
43.12 ± 2.4%
50 μg/mL curcumin
4T1
CUR-HSA-DOPE NPs
25.2 ± 5.8% and 11.9 ± 8.6%
free CUR
34.5 ± 6.6% and 48.3 ± 7.2%
50 μg of curcumin
Immunofluorescence TUNEL assay
↑ Apotosis CUR-HSA-DOPE NPs
None
Kumari et al., 2016 [54]
India
MDA-MB-231
Human
Curcumin at 50 μg/mL
Curcumin
and curcumin-loaded nanoparticles (curcumin in mPEG-PLA micelles) (CUR-HSA-DOPE NPs)
24 hNot reportedMTT Assay
CUR-mPEG-PLA231 35.1 ± 8.5
free CUR
65.7 ± 1.0%
50 μg/mL
Not reportedNone
Laha et al., 2018 [55]
India and USA
MDA-MB-468
Human
Curcumin at
20,
40, 60, 80, 100, and 120 mM
12 and 24 hNot reportedNot reportedAnnexin V-FITC staining
Apoptotic cells: 25% and 91%.
None
Lai et al., 2012 [56]
China
MCF-7,
BT-474,
MDA-MB-231, and normal breast cells
Human
Curcumin at
10 μg/mL
72 hColorimetric analysis of sulforhodamine B
↓ cell proliferation (MCF-7, BT-474, and MDA-MB-231 cells).
Not reportedNot reportedNone
Li et al., 2018 [3]
China
MDA-MB-231
Human
Curcumin at 10 g/mL
Nanostructured platform
curcumin and
curcumin nanoparticle
MSN/IR780-PEI-FA
160 mg/kg
24 and 48 hNot reportedNot reportedFlow cytometry
CUR and free MSN/CUR induced the G2/M
phase of the cell cycle.
Liu et al., 2013 [58]
China
4T1
Mouse
Curcumin at 100 μg/mL
Nanostructured platform
Nanoparticle with self-assembled polymeric micelles (CUR-M) loaded with curcumin (CUR)
48 hNot reportedMTT assay
Both CUR-M and Free CUR drastically inhibited cell growth in a dose-dependent manner.
TUNEL assay by immunofluorescence staining
Apoptotic index
CUR-M: 15.77 ± 2.74%,
Free CUR: 9.42 ± 2.13%
p < 0.001)
None
Liu et al., 2009 [58]
China
MDA-MB-231
Human
Curcumin at
1, 1.25, 2.5, 5,
10, and 20 mg/mL
24 and 48 hMethod (NR)
Inhibition of cell growth by 60–70% with
1.25 mg/mL curcumin.
Inhibition of cell growth by 50–60% with
2.5 mg/mL curcumin.
Not reportedNot reportedNR
Lv et al., 2014 [61]
China
MDA-MB-231
Human
Curcumin at
1–100 μL
24 and 48 hNot reportedMTT assay
↓ significant reduction in the number of viable cells in a time- and dose-dependent manner.
Flow cytometry of fixed nuclei
↑ in the number of apoptotic cells in a dose-dependent manner.
None
Masuelli et al., 2013 [63]
Italy
MDA-MB-231,
MDA -MB-435,
MDA-MB-453,
MDA-MB-468,
T-47D,
MCF7, BT-474,
SK-BR-3
Human
Mammary cancer cells (H-2”) (TUBO)
Humanized mouse
Mammary cancer cells (H-2”) (TUBO)
Mouse
Curcumin
6 to 50 pM
24 and 48 hNot reportedNot reportedPro-apoptotic Bax and anti-apoptotic Bcl-2 expression
CUR induced apoptosis in all investigated cell types.
None
Mehta et al., 1997 [64]
USA
MCF7
Human
Curcumin
1 to 3 μg/mL
72 h[3H]thymidine incorporation and flow cytometry.
Cell growth inhibition in a time- and dose-dependent manner, correlated with the inhibition of ornithine decarboxylase activity.
Not reportedFlow cytometry
Curcumin-induced cell death was not due to apoptosis or any significant change in the expression of apoptosis-related genes, including the Bcl-2, p53, cyclin B, and transglutaminase genes.
NR
Montazeri et al., 2017 [65]
Iran
MCF7
Human
Curcumin at
23, 17, and 14 µM
Dendrosomal curcumin
(DNC) for 48 h (28–35 μM) and 72 h (23–25 μM)
24, 48, and 72 hNot reportedNot reportedFlow cytometry
Total apoptosis by DNC:
24 h: 30.34 ± 0.011%
48 h: 33.83 ± 0.005% 72 h: 61.83 ± 0.009%
None
Mukhopadhyay et al., 2020 [67]
India
MDA-MB-231
Human
5 mg of
curcumin
Nanostructured platform
Polymeric NPs PLGA/PVA with or without folate (F)
24 hNot reportedNot reportedFlow cytometry
Apoptosis
CUR-NP-F: 29%
Free CUR: 20%
Sarighieh et al., 2020 [70]
Irã
MCF7
Human
Curcumin
5, 10, 20, 40, 80, and 160 μM
24 hNot reportedMTT assay
Curcumin decreased the cell viability of MCF-7 cells.
Flow cytometry
Apoptosis
24.6%
None
Sun Shih-Han et al., 2012 [73]
Taiwan
MDA-MB-231/Her2
Human
Curcumin at
30 and 50 mM
24 hNot reportedNot reportedFlow cytometry
Apoptosis
occurred at a higher dosage (50 mM).
None
Sun Xiao-Dong et al., 2012 [74]
China
MDA-MB-231
Human
Curcumin at
10, 20, and 30 μmol/mL
48 hMTT assay
The inhibitory effect on MDA-MB-231 cell proliferation peaked upon treatment with 30 μmol/mL curcumin (p < 0.01).
Not reportedFlow cytometry
Apoptosis control 2.76% and
Curcumin 26.34%,
30 μmol/mL
(p < 0.01).
None
Wang Xet al., 2017 [76]
China
MCF-7
Human
Curcumin [0 (with DMSO vehicle), 0.5, 1.0, 2.0, 5.0, and 10.0 µM]24, 48, and 72 hMTT assay
↓ cell growth (treatment with 0, 0.5, 1.0, 2.0, 5.0, and 10.0 µM curcumin).
Not reportedFlow cytometry
Apoptotic cell death within 48 h
upon treatment with 2 µM (p = 0.0021) and 5 µM (p = 0.0004) curcumin.
None
Yang et al., 2017 a [76] ChinaMCF-7
Human
Curcumin at
50 μm
Nanostructured platform
Micelle NPs (PPBV triblock copolymer)
24 hNot reportedNot reportedFlow cytometry
Apoptotic cell death
Younesian et al., 2017 [80]
Irã
SKBR3
Human
Curcumin at 2.5, 10, 15, 20, 25, and 30 μM24, 48, and 72 hNot reportedNot reportedFlow cytometry
Apoptosis: 4.37% with 0 μM, 27.46% with 5 μM, 64.98% with 10 μM, 75.90% with 15 μM, and 76.92% with 20 μM curcumin.
None
Yu et al., 2021 [81]
China
4T1
Mouse
Curcumin at
5, 10, and 15 μM
24 hNot reportedMTT assay
↓ of cell viability by 16% using 15 μg/mL curcumin
Not reportedNone
Zong et al., 2012 [84]
China
MCF-7
Human
Curcumin at 10, 20, 50, and 100 μM48 hMTT assay
↓ cell growth by 37%, 54%, and 73% using 20, 50, and 100 μM curcumin, respectively.
Not reportedNot reportedNone
MTT assay, MTT Assay Protocol for Cell Viability and Proliferation, ↓: inhibition, ↑: activation.
Table 2. Characteristics of the studies conducted on experimental animal models included in the systematic review on curcumin and breast cancer.
Table 2. Characteristics of the studies conducted on experimental animal models included in the systematic review on curcumin and breast cancer.
Author/Year/CountryExperimental Animal Model *InterventionOutcomeConflicts of InterestEthical Approval
Treatment
Follow-Up
Dose (mg/kg)/Administration RouteAnti-Tumor Activity (Size or Volume of the Tumor)
Abd-Ellatef et al., 2020 [38]
Italy and Egypt
Balb/c/n = 8/JC/mouse/(1 × 107 cells)/mammary fat padVT: 50 mm3; three times (on days 1, 7, and 14); vehicle-free CUR:
10% DMSO suspension v/v
Follow-up: 18 days
Nanostructured platform
Solid lipid nanoparticles (SLNs) with or without chitosan (CS) coating (cholesterol; trilaurin, butyl lactate, Epikuron® 200, Cremophor® RH60, sodium taurocholate, Pluronic® F68)
5 mg/kg; Intravenous administrationCURC-CS-SLN and CURC ↓ VT (35%);
Free CUR: no VT ↓; p < 0.01
NoneYes
Alizadeh et al., 2015 [29]
Iran
Balb/c/n = 8/Transplantation of spontaneous mouse mammary tumor/pieces < 0.3 cm3/subcutaneous administration in the left flank14 days after tumor induction; daily for 24 days
Follow-up: 35 days
Nanostructured platform
Micelles/polymersomes NPs (PNP) [monomethoxyPEG (mPEG 2000), oleic acid (OA)]
Dose: (NR); Intraperitoneal administrationCUR-NP ↓ VT (80%); p < 0.05NoneYes
Bansal et al., 2014 [30]
USA
Female ACI mice/
5 to 6 weeks old/mammary tumorigenesis mediated by 17β-estradiol (E2)/9 mg of E2/back
4 days after tumor induction/
Curcumin implants (n = 6)
Curcumin diet (n = 6)
Follow-up: 6 months
Curcumin 1000 ppm
via diet
Two 2 cm implants, 200 mg/implant, 20% p/p drug load
10.9 mg of curcumin for 25 days
subcutaneous administration
Curcumin implant: ↓ VT (35%)
Curcumin administration via diet: ineffective
NoneYes
Bimonte et al., 2015 [7]
Italy
Foxn1 nu/nu female mice/n = 16, 6-to-8-week-old/human breast cancer cell line MDA.MB231/2.5 × 106 cells/right flankAfter reaching 30–60 mm3, normal diet (n = 8) and
diet containing 0.6% curcumin were administered (n = 8).
Follow-up: 6 weeks
0.6% Curcumin administration
via diet
↓ VT (% NR) (p = 0.0195)NoneNR
Chen et al., 2017 [32]
China
Balb/c/n = 5/BT-549/human (2 × 106 cells)/subcutaneous administration in the right upper thigh200 mm3 VT 35 mg/kg;
Fourteen days, every 2 days Intratumoral—Vehicle
Free CUR: NR
Follow-up: 30 days
Nanostructured platform
Micelle NPs [POCA4C6 (phosphorylated calixarene) micelles—PM]
5 mg/kg;
Intratumoral administration
CUR-NP ↓ VT (60%);
Free CUR: ↓ VT (34%);
p < 0.05
NoneYes
Ghosh et al., 2021 [39]
India
Swiss albino mice/3 groups (n = 5)/MCF-7 and MDA-MB 231 cells (human)/veinAlternating days after tumor induction
Follow-up: 2 weeks
Nanostructured platform
Nanoparticles: MSN-Curcumin (MSN-C) and MSN-Hyaluronic acid-Curcumin (MSN-HA-C)
10 mg/kg;
intravenous administration
MSN-HA-C
↓ VT (% NR); p < 0.05
NoneYes
Greish et al., 2018 [40] BahrainBalb/c/n = 5/4T1/mouse/(1 × 106 cells)/bilaterally on flanksVT: 100 mm3; frequency of treatment: unclear; Treatment: 10 days
Follow-up: 9 days
Nanostructured platform
Micelles (curcumin-metal complex and SMA)
10 and 20 mg/kg; Intravenous administrationCUR-NP-10 mg/kg ↓ VT (61%);
CUR-NP-20 mg/kg ↓ VT (92%); p < 0.05
NoneNR
Grill et al., 2018 [41]
Estados Unidos
Balb-neuT mice/n = NR/HER-2-positive
breast cancer cells/ten breast pads
At 2, 4, 7, or 12 weeks of age, and once a month thereafter
Follow-up: 24 weeks
Nanostructured platform
Curcumin-loaded microparticles
Curcumin (20 mg) and PLGA (20 mg)
140 mg of microparticles, corresponding to 58.2 mg of curcumin/administered via subcutaneous injectionCurcumin MP ↓ VT (60%); p < 0.05NoneYes
Hashemzehi et al., 2018 [42]
Iran
Balb mice/n = 4/MCF-7 cells (human)/flanksVT: 100 mm3; 7 days after tumor induction
Follow-up: 22 days
Nanostructured platform
Nanocurcumin—phytosomal curcumin
Dose: (NR); NRCurcumin
↓ VT (22.2%)
Curcumin + 5-FU
↓ VT (53.3%)
NoneYes
He et al., 2019 [43]
China
Balb/c/n = 6/4T1/mouse/(1 × 106 cells)/subcutaneous administration in right backVT: 100 mm3
Every 4 days for 4 times
Free CUR: (NM)
Follow-up: 21 days
Nanostructured platform
Polymeric micellar NPs [amphiphilic diblock copolymer—mPEG-b-PLG (Se)-TP]
5 mg/kg;
Intravenous administration
CUR-NP ↓ VT (62.9%);
Free CUR: ↓ VT (55%); p < 0.05
NoneYes
Huang et al., 2020 [46] ChinaBalb/c/n = 5/4T1/mouse/NR/Flank miceVT: 40–50 mm3/every 2 days for 5 times
Follow-up: 16 days
Nanostructured platform
50 mg/kg;
Intravenous
CUR-NP ↓ VT (38%); p < 0.05NoneYes
Ji et al., 2020 [47]
China
Balb/c/n = 5/4T1/mouse/(1 × 106 cells)/subcutaneous administration in the right flankPolymeric NPs (HA-CHEMS); pH-sensitive
First day of treatment: NR;
Every 2 days
Vehicle-free CUR: (NM)
Follow-up: 10 days
Nanostructured platform
Nanocrystal NPs with or without HA
5 mg/kg; IntravenousHA@CUR-NCs ↓ VT (86%); CUR-NP ↓ VT (39%); Free CUR: ↓ VT (21%); p < 0.05NoneYes
Jin et al., 2017 [49]
China and USA
Balb/c nude rats/n = 5/MCF-7/human/(1 × 107 cells)/subcutaneous administration in the dorsal flank7 days after tumor induction;
every 24 h for 20 times
Free CUR: NR
Follow-up: 3 weeks
Nanostructured platform
Polymeric NPs with or without EGFR-targeting peptides (GE11) (PLGA-PEG);
5 mg/kg; Intravenous administrationCUR-NP-GE11 and CUR-NP ↓ VT (80%);
Free CUR: sem VT ↓; p < 0.05
NoneYes
Jung et al., 2018 [50] República da CoréiaBalb/c nude rats/n = 4/MDA-MB-468 cells/human/(5 × 106 cells)/right shoulderVT: 50 mm3; three times a week; eight injections in all
Follow-up: NR
Nanostructured platform
CUR-NP e EGF-CUR-NP
10 mg/kg; Intraperitoneal administrationCUR-NP-EGFR ↓ VT (59.1%);
CUR-NP no ↓ VT; p < 0.05
NoneYes
Kumari et al., 2020 [52]
India
Balb/c mice/n = 18/Mouse (4T1)/50 μL, 1 × 106 cells/subcutaneous administration in
left flank
VT: 50 mm3;
Follow-up: 21 days Nanostructured platform
CUR treatment (Free CUR group (0–24 μg/mL)) and CUR-HSA-DOPE NPs treatment (CUR-HSA-DOPE group)
25 mg/kg;
Intravenous administration
CUR-HSA-DOPE ↑ VT (80.41%);
Free CUR ↑ VT (86.30%)
NoneYes
Laha et al., 2018 [55]
India and USA
Balb/c/n = 6/4T1/mouse/NR/mammary fat pad10 days after tumor induction; every 5 days for four times
Follow-up: 20 days
Nanostructured platform
Metal organic frameworks NPs (IRMOF-3) with or without folic acid (FA) [(Zn(NO3)2; NH2-H2 BDC]
2 mg/kg (* unclear);
Route of administration: (NM)
CUR-NP-FA ↓ VT (61%); CUR-NP ↓ VT (44%); p < 0.05NoneYes
Lai et al., 2012 [56]
Taiwan
Nude mice/n = 16/BT-474 cells overexpressing HER-2 (1 × 107)/right flank subcutaneous route of administration21–28 days after xenograft inoculation. VT:50–100 mm3
Follow-up: after 4 weeks
45 mg/kg curcumin
injected
intra-peritoneally
Herceptin and curcumin VT 34.1 ± 25.0 mm3
Curcumin
VT 63.6 ± 25.7 mm3
p = 0.079
Li et al., 2018 [3]
China
Balb/c/n = 4/MDA-MB-231/human/(1 × 107 cells)/subcutaneous administrationTumor diameter: 4 mm; every 3 days for six times in all
Free CUR: NR
Follow-up: 18 days
Nanostructured platform
Mesoporous silica nanoparticles with hyaluronan (MSN-HA) or polyethyleneimine-folic acid (MSN-PEI-FA).
8 mg/kg; Intravenous administrationCUR-NP-PEI-HA ↓ VT (50%);
Free CUR: no VT ↓; p < 0.01
NoneYes
Lin et al., 2016 [57] ChinaBalb/c nude mice/n = 6/MCF-7/ human/(NM)/Subcutaneous administration in the right axillaFirst day of treatment: NR once every 3 days for 15 days
Vehicle-Free CUR: (NM)
Follow-up: 15 days
Nanostructured platform
Lipid-based NPs (NLC) with or without folate coating (FA) (PEG-DSPE, soy lecithin, castor oil, Tween 80, and Precirol ATO-5)
Dose: NR;
Intravenous administration
CUR-NP-FA ↓ VT (~83%); CUR-NP ↓ VT (~66%); Free CUR: ↓ VT (31%)NoneNR
Liu et al., 2013 [58]
China
Balb/c mice n = 12; 6 per group/4T1/5 × 105 cells/right flank/subcutaneous administrationFrom day 4, palpable tumors were daily injected with the treatment agent
intravenously for 10 days
Follow-up: 25 days
Nanostructured platform
Self-assembled polymeric micelles (CUR-M) loaded with curcumin (CUR)
CUR-M
(30 mg/kg body weight)
Free CUR (30 mg/kg body weight)
CUR-M ↓ VT (68%);
p < 0.01
Free CUR: sem ↓ VT (35%)
NoneYes
Lv et al., 2014 [61]
China
Balb/c nude mice/n = 8 per group /MCF-7 and MDA-MB-231/2 × 106 cells/subcutaneous administration in the backAfter reaching 60 mm3/treatment days alternating
Follow-up: 4 weeks
Curcumin
50 µg/kg, 200 µg/kg
Intraperitoneal injections
Cur 50 µg/kg ↓ VT (54%); p < 0.05
Cur 200 µg/kg ↓ (73%); p < 0.05
VT
NoneYes
Lv et al., 2015 [60]
China
Kunming mice/n = 6/EMT6/mouse/(1.0 × 107 cells/mL)/Subcutaneous administrationVT: 300 mm3; daily for 9 days
Vehicle-free CUR: cremophor EL:dehydrated alcohol (1:1, v/v) and diluted with saline solution
Follow-up: 14 days
Nanostructured platform
Polymeric NPs (PEG-PCDA) with or without biotin
10 mg/kg; Intravenous administrationCUR-NP ↓ VT (69%); CUR-NP-biotin ↓ VT (79%);
Free CUR: ↓ TV (32%); p < 0.05
NRYes
Mahalunkar et al., 2019 [62]
India,
Germany and Norway
Balb/c/n = 6/4T1/mouse/(1 × 105 cells)/Mammary fat padFirst day of treatment: (NM) Twice a week for 2 weeks
Vehicle-free CUR: (NM)
Follow-up: 21 days
Nanostructured platform
Metallic gold NPs (CurAu-PVP) with folic acid (FA) (HAuCl4 and PVP polymer)
10 mg/kg; Intratumoral administrationCUR-NP-FA ↓ VT (51%);
Free CUR: no ↓ VT; p < 0.006
NoneYes
Masuelli et al., 2013 [63]
Italy
Transgenic BALB-neuT mouse/n = 5 per group /NRAfter the diameter reached 15 mm,
CUR (2 mg in 50 |.il oil with),
with oil (50 |.il) or water (50 |.il) was administered three times a week.
Follow-up: 30 weeks
Curcumin
6–50 µM
Oral administration
No
mice treated with CUR exhibited
tumor growth
at week 22,
(p < 0.01).
Cur ↓ VT (52%) (p < 0.05)
NoneNR
Mukerjee et al., 2016 [66] USABalb/c nude rats/n = 8/MCF10CA1a/human/(3 × 106 cells)/flankVT: 70 mm3; Three times a week for 30 days
Follow-up: 32 days
Nanostructured platform
Polymeric NPs [PLGA/PVA with or without antibody targeting (AnxA2)]
20 mg/kg; Intravenous administrationCUR-NP-AnxA2 ↓ VT (44.0%); CUR-NP ↓ VT (33.5%); p < 0.05
CUR-NP-AnxA2 ↓ PT (53.0%); CUR-NP ↓ PT (30%); p < 0.05
NRNR
Mukhopadhyay et al.,
2020 [67]
India
Balb/c nude rat/n = 5/MDA-MB-231/human/(5 × 106 cells)/Right flank8 days after induction; three times a week
Follow-up: 29 days
Nanostructured platform
Polymeric NPs [PLGA/PVA with or without folate (F)]
20 mg/kg
Route of administration: unclear
CUR-NP-F ↓ VT (90%); CUR-NP ↓ VT (75%); p < 0.05NRYes
Pal et al., 2019 [68]
India
Balb/c mice/n = 5 per group /human MCF-7,
MDA-MB-231, MDA-MB-468, and murine
4T1/100 L/abdominal skin
Treatment for 20 days at 3-day intervals after 10 days of
tumor implantation
Follow-up: 30 days
Nanostructured platform
Synthesis of curcumin-loaded microsphere
(10% by weight polymer)
PLGA@CCM@FA
2000 µg/kg
Route of administration: unclear
PLGA—VT 0.092 mm3 ↓ VT (25%)
PLGA @ CCM—VT 0.064 mm3 ↓ VT (48%)
PLGA @ CCM @FA-VT—0.031 mm3 ↓ VT (75%)
NRNR
Sahne et al., 2019 [69] IrãBalb/c/n = 4/4T1/mouse/NR/ssubcutaneous administration in the flankVT: 50–100 mm3; daily
follow-up: 3 weeks
Nanostructured platform
Graphene oxide NPs (GO NPs with CMC, PVP, PEG, and FA)
4 mg/kg; Intravenous administrationCUR-NP-FA ↓ VT (86%); p < 0.05NoneYes
Shiri et al., 2015 [71]
Irã
Balb/c/n = 9/4T1/mouse/(1 × 106 cells)/left flankThird day after tumor induction
Follow-up: 35 days
Nanostructured platform
Dendrosome NPs (DNC) [composition: not mentioned (patent number: 71753)].
40 or 80 mg/kg
Route of administration: NR
NP-40 mg/kg ↓ VT (72%); NP-80 mg/kg ↓ VT (76%); p < 0.05
NP-40 mg/kg ↓ VT (61%); NP-80 mg/kg ↓ VT (64%); p < 0.05
NRYes
Shukla et al., 2017 [72]
India
Balb/c mice/n = 3/(1 × 106 cells)/subcutaneous administration in hind skin10 days from tumor inoculation; daily administration for 28 days: gum acacia (1%, w/v).
Follow-up: 42 days
Nanostructured platform
Lipid-based CPC-SNEDDS NPs (Phospholipid, castor oil, Tween 80, and PEG 400)
100 mg/kg; oral1) CUR-NP ↓ VT (58.9%);
Free CUR ↓ VT (29.5%); p < 0.001
NoneYes
Vakilinezhad et al., 2019 [75]
Irã
Sprague–Dawley rats/n = 6/Chemically-induced mammary tumors (MNU)4 months after tumor induction; Once a week for 4 weeks
Free curing vehicle: aqueous suspension
Follow-up: 20 weeks
Nanostructured platform
Polymeric NPs (PLGA-PVA)
2.5 mg;
Intravenous
CUR-NP ↓ VT (20%); Free CUR: ↓ VT (16%); p < 0.05NoneYes
Wang et al., 2018 [77] ChinaNude mice/n = (NM)/MDA-MB-231/human/(1.5 × 106 cells)/subcutaneous2 months after tumor induction;
daily
Free CUR: (NM)
Follow-up: 2 weeks
Nanostructured platform
Polymeric NPs (MPEG-PCL)
1 × 10−3 M;
Intravenous administration
CUR-NP ↓ VT (82%); Free CUR: ↓ VT (49%); p < 0.01NoneYes
Yang et al., 2017 a [78] ChinaBalb/c nude mice/n = 5 MCF-7/human/(1 × 107 cells)/subcutaneous administration in the flankVT: 200 mm3
Every other day, five times; total duration: 20 days
Free CUR vehicle: NR
Follow-up: 20 days
Nanostructured platform
Hybrid NPs [PLGA NPs coated with a modified hyaluronic acid (HA hybrid)]
15 mg/kg; IntravenousHA-Hybrid NPs/CUR ↓ VT (43.8%, day 12); ↓ VT (24%, day 20); p < 0.05NRYes
Yang et al., 2017 b [79] ChinaBalb/c nude mice/n = 5 MCF-7/human/(1 × 107 cells)/subcutaneous administration in the flankVT: 200 mm3
Every other day, five times; total duration: 20 days
Free CUR vehicle: NR
Follow-up: 20 days
Nanostructured platform
Micelle NPs (PPBV triblock copolymer)
10 mg/kg; IntravenousPPBV micelles/CUR ↓ VT (58.5%, day 12);
↓ VT (28.9%, day 20); p < 0.05
NRYes
Yu et al., 2014 [82] ChinaBalb/c nude mice/n = 5/MCF-7/human/(3 × 106 cells)/subcutaneous administration in the right flankVT: 100–400 mm3; Every other day for 5 times for 24 days in all
Follow-up: 25 days
Nanostructured platform
Micelle NPs (MPEG-PLA with or without PAE)
40 mg/kg;
Intravenous administration
CUR-NP-PAE ↓ VT (65.6%); CUR-NP ↓ VT (47.1%); p < 0.05NRYes
Yu et al., 2021 [81]
China
Balb/c mice/ murine 4T1/NR/intradermal administration in the back of the neckVT: 150–200 mm3, administration via tail vein every 3 days; 14 days in all
Follow-up: 16 days
Nanostructured platform
curcumin (CUR), zeolitic imidazolate framework-8 nanoparticles (ZIF-8), and hyaluronic acid (HA)
CUR@ZIF-8 19.6 mg of
CUR@ZIF-8@ HA 20.9 mg
Intravenous administration
CUR@ZIF-8 ↓ VT (12.5%);
CUR@ZIF-8@HA ↓ VT (62.5%);
NoneYes
Yuan et al., 2018 [83] ChinaBalb/c nude mice/n = 6/MCF-7/human/(3 × 106 cells)/right flankVT: 100 mm3; every other day, four times
Follow-up: 18 days
Nanostructured platform
Polymeric NPs (mPEG-PLGA-Pglu)
2.5 mg/kg; intravenous administrationCUR-NP ↓ VT (28.0%); p < 0.05
CUR-NP ↓ PT (22.5%); p < 0.05
NoneYes
* Animal type/sample size/injected cell type/source/cell concentration/cell insertion site; NR: not reported; VT, tumor volume; ↓: inhibition; ↑: activation.

3.2.3. Cell Viability

Cytotoxicity in breast cancer cell lineages was assessed using the MTT assay. Curcumin significantly decreased the viability of MCF-7 malignant cells in a time and dose-dependent manner [27,49,70]. In another trial, a decrease in the viability of MCF-7 cells by 49% and of MDA-MB-453 cell cultures by 48% following treatment with 20 µM curcumin for 24 h was observed [36], while another study reported that curcumin did not affect the viability of MCF-7 cell cultures [51]. Cells were treated with 1, 5, 10, 30, and 50 μM curcumin for 24 h at 37 °C.
There was a significant decrease in the viability of MDA-MB-468 cells upon treatment with 10 µM curcumin [50]. The viability of triple-negative MDA-MB-231 cell cultures reduced by up to 25% upon treatment with 15–100 μM curcumin for 24 h [28,37,53,54,61]. There was a 55.2% reduction in the viability of MDA-MB-23 colonies upon treatment with 50 μg/mL curcumin [52]. In T47D cell lineages, viability reduced by 48% and 60% upon treatment with 20 µM curcumin [35].
Mouse 4T1 cultures showed a significant reduction in cell viability upon treatment with pure 6–50 pM curcumin [43,53,81]. Curcumin CUR-M and free CUR nanoparticles also inhibited cell growth in a dose-dependent manner [58].

3.2.4. Apoptosis and/or Interruption of Cell Cycle

In most studies, apoptosis and/or interruption of the cell cycle were assessed using the MTT assay [28,36,37,39], quantitative image analysis [34], Annexin-V/PI double staining [35,48], immunofluorescence TUNEL assay [53,58], Annexin V-FITC staining [55], pro-apoptotic Bax and anti-apoptotic Bcl-2 expression evaluation [63], and flow cytometry.
In the breast cancer MCF-7 cell lineage, apoptosis occurred in 4.6% of the cells upon treatment with 25 μM curcumin [34], and in 28.7% and 49% of the cells upon treatment with 30 μM curcumin [31,36,48]. Other studies reported 14.9% apoptosis in MCF-7 colonies treated with 10 µg/mL curcumin delivered via nanoparticles [49]. There was 24.6% apoptosis in MCF-7 cells incubated under normoxic and hypoxic conditions for 24 h and treated with curcumin at different concentrations (5, 10, 20, 40, 80, and 160 μM) [70]. Wang et al. also reported apoptosis following treatment with 2 and 5 µM curcumin for 48 h [76].
In the triple-negative MDA MB-468 cell lineage, the apoptosis frequency was 25% and 91% after treatment for 12 and 24 h, respectively [55]. In addition, 30 μM curcumin induced apoptosis in 31.36% of MCF-7/LCC2 cells [48] and 34.70% of LCC9 cells [48]. Other studies also reported apoptosis in triple-negative MDA-MB-231 cells treated with 10, 12, 20, 30, and 50 μM curcumin for 24 and 48 h [3,7,28,31,33,37,39,47,53,61,67,74], and in colonies of SK-BR-3 cells treated with 5, 10, 15, and 20 μM curcumin [63,80]. In T47D cells, 30 µM curcumin induced 10.9% apoptosis in 24 h, 5.2% apoptosis in 48 h [35], and 30.09% apoptosis in 72 h [44].
Mouse 4T1 cell lines showed increased apoptosis in response to treatment with 6 to 50 pM curcumin [43,53,63]. Moreover, CUR-NPs at 0–100 μg/mL also induced apoptosis in a dose-responsive manner [58].

3.2.5. Animal Studies

Thirty-seven studies on animal models met the inclusion criteria [3,7,29,30,32,38,39,40,41,42,43,46,47,49,50,53,55,56,57,58,60,61,62,63,66,67,68,69,71,72,75,77,78,79,81,82,83]. Curcumin was delivered via diet in two studies [7,63], diet and implant in one study [30], intraperitoneal injection in two studies [56,61], and different modes using nanoparticles in 32 studies. The results of these studies are listed in Table 2, with the animal species, sampling size, type of cells injected, cell concentration, cell insertion site, treatment, follow-up, dose, and route of administration specified. The studies were heterogeneous with respect to the animal model, follow-up, curcumin dose, and route of administration.
A curcumin-encapsulated polymer micelle formulation was developed showing antitumor and anti-metastatic activities in breast cancer cells [58]. Micelles loaded with curcumin inhibited tumor activity and induced minimal collateral effects in vivo compared to a free curcumin formulation (free-CUR) [43]. Reduction in tumor volume increased significantly following treatment with CUR-NPs (20–92%) [29,32,38,40,46,57,58,60,67,72,75,76,83] rather than with free CUR (0–55%) [32,38,40,43,47,49,60,62,72,75,77,83].
Other curcumin delivery methods and their corresponding tumor magnitude percentile reductions were as follows: HA@CUR-NCs (86%) [47], CUR-NP-biotin (79%) [60], curcumin + 5-FU (53.3%) [42], CUR-NP- AnxA2 (44.0%) [66], CUR-NP-PEI-HA (50%) [3], HA-Hybrid NPs/CUR (43.8%) [78], PPBV micelles/CUR (58.5%) [79], CUR-NP-FA (51–86%) [57,62,69], CUR-NP-PAE (65.6%) [82], CUR@ZIF-8@HA (62.5%) [81], and CUR-NP-EGFR (59.1%) [55]. Furthermore, the synthesized nano-hybrid MSN-HA-C increased anticancer efficacy when compared to Free CUR [39].
Intracellularly degradable, self-assembled amphiphilic biotin-poly (ethylene glycol)-b-poly (curcumin–dithiodipropionic acid) nanoparticles exhibited excellent anticancer activity in vivo due to their high tumor-targeted accumulation and stimuli-triggered intracellular drug release [60]. Moreover, these nanoparticles could be loaded with other anticancer drugs, which could promote synergistic oncologic effects in vivo [60].
In another trial, compared to control PLGA microparticles, curcumin-loaded microparticles retarded oncogenesis in a Balb-neuT transgenic mouse model. PLGA microparticles accelerated oncogenesis compared to a saline control. This unanticipated collateral effect of PLGA microparticles may be related to the high dose of microparticles for optimal in vivo concentration of curcumin [41].

3.3. Conflict of Interest and Ethics Committee Approval

Only six studies were approved by their respective ethics committees on animal use [7,40,57,63,66,68], while there was no mention of potential conflicts of interest in eight studies [60,66,67,68,71,78,79,82]. The authors of the remaining articles declared no conflicts of interest.

3.4. Overall Quality of Evidence

Thirty-nine studies were rated as moderate with respect to quality of evidence using the GRADE approach [25], as shown in (Table S3). These studies were not representative of the results of all assessed outcomes.
The evaluation of the risk of bias based on the SYRCLE RoB Toll guidelines for animal model studies is shown in Table 3. Most studies did not clearly state information on assignment, randomization, and blinding, which are critical aspects for assessing the quality of evidence.

4. Discussion

This systematic review highlighted some of the promising antitumor activities of curcumin reported in in vitro studies, as well as its potential for tumor volume reduction in animal models. At different concentrations, curcumin inhibited cell proliferation, reduced cell viability, and induced apoptosis in several human and animal breast cancer cell subtypes. In vivo data showed that curcumin reduced tumor volume in human and murine mammary cells when administered either orally, via implants, or via intraperitoneal injection, or when delivered via different curcumin nanoparticle formulations.
In vitro studies showed inhibitory activity of curcumin on cell proliferation, induction of cell viability, and apoptosis at different concentrations. The anti-proliferative effect of curcumin was attributed to its regulatory effects on protein kinases, the cell cycle, and transcription factors, including NF-κB [85]. Curcumin significantly inhibited the growth of MDA-MB-231 and MCF-7 human breast cancer cells by inducing apoptosis in a gradual, dose-dependent method, which was related the increase in the Bax/Bcl-2 ratio [34,61].
The cell cycle is divided into four phases: G1, S, G2, and M [85]. Dendrosomal curcumin increases the number of cells in the SubG1 phase and reduces the number of cells in the G1, S, and G2/M phases [65]. Early-stage apoptosis showed the inhibition of cell growth through the early phase. Real-time PCR revealed a gradual increase in the mRNA levels of BAX, NOXA, and p21, with a decrease in Bcl-2 expression [65]. The magnitude of anticancer effects and induction of apoptosis are essential for investigating antineoplastic therapy. Apoptosis occurred via intrinsic or mitochondrial pathways [85]. Apoptotic pathways were modulated via NF-κB and Bax [39,67]. Curcumin was also shown to downregulate the expression of cyclin D1, PECAM-1, and p65, which are regulated by NF-κB [7,35]. Figure 2 shows different mechanisms of action of curcumin in breast cancer, including cell proliferation, cell viability, and apoptosis.
The PLGA@CCM@FA nanoparticle formulation triggered apoptosis in human triple-negative breast cancer cells by positively regulating cleaved caspase-3 and downregulating p-AKT expression [68]. Curcumin also induced caspase-mediated apoptosis by activating the expression of polyamine catabolic enzymes, with the subsequent generation of toxic molecules such as H2O2 in MCF-7, MDA-MB-453, and MDA-MB-231 GH+ breast cancer cells [35]. Curcumin-encapsulated polymeric micelles should be considered for breast cancer treatment, as they reduced the proliferation of breast cancer cells [58]. Curcumin-loaded micelles also showed significant tumor-inhibiting properties as well as minimal in vivo collateral effects compared to free-CUR formulations [43]. A study revealed that alginate–chitosan hydrogel loaded with curcumin significantly reduced the viability and induced the apoptosis of malignant cells. Therefore, this system presents promising anticancer drug delivery properties [86].
Conversely, one of the pharmacological limitations of orally administered curcumin is its low bioavailability owing to its low solubility in water and rapid metabolism, which may hinder its clinical application [72,87]. In a randomized clinical study, water-soluble injection formulations of curcumin for parenteral/intravenous administration showed up to 100% bioavailability, demonstrating its potential clinical application [87]. Moreover, a liquid droplet nanomicellar formulation containing Gelucire® and polysorbate 20 (BioCurc®) showed optimal bioavailability, with more than 400-fold greater absorption than non-formulated curcumin [88].
This study highlights different curcumin nanoparticle formulations with optimal bioavailability, causing substantial mammary tumor-reducing effects. Recent advances in micro-and nanoformulations of curcumin with enhanced absorption yield helped improve the serum levels of the active components. These formulations have a wide range of potential applications and properties, including tissue protection [89].
The results discussed in this review support randomized clinical investigations of the antitumor properties of curcumin in patients with breast cancer. Considering the diversity and heterogeneity of breast cancer subtypes, further studies will provide deeper insights into the effects of curcumin on specific types of mammary neoplasms to determine the effects on tumor markers, metastasis, and patient outcomes. Moreover, the efficiency and safety of curcumin in combination with other chemotherapeutic drugs should be established. In future clinical trials, tumor characteristics should be considered to support clinical decision-making. Both human patients and animals showing mammary neoplasms may benefit from curcumin-based therapies in the near future, as indicated by evidence from studies on animal models.
Although eight ongoing clinical assays on the effects of curcumin on breast cancer have been registered on clinicaltrials.gov to date, to the best of our knowledge, only one randomized controlled double-blinded clinical trial has been published [87]. In the said study, 150 women with advanced metastatic breast cancer were randomly assigned to receive paclitaxel chemotherapy (80 mg/m2) plus placebo or paclitaxel with curcumin (CUC-1® solution, 300 mg, administered intravenously once a week) for 12 weeks, with three months of follow-up. The paclitaxel–curcumin combination provided a superior objective response and physical performance after two weeks of treatment. Intravenous curcumin was safe, did not negatively affect the patients’ quality of life, and decreased fatigue [87].
Currently, the data available only pertain to a trial at an advanced stage; therefore, studies focusing on early stages and, in particular, net-adjuvant chemotherapy are lacking. Addressing this knowledge gap remains essential. There are good prospects for the use of curcumin in cancer management, although its clinical development is limited due to its low bioavailability and aqueous solubility [90]; however, efforts have been made to improve the solubility, stability, and bioavailability of curcumin. For example, one strategy employed to obtain curcumin derivatives is chemical modification or synthesis of their analogues. Furthermore, curcumin encapsulated in protein nanoparticles demonstrated improved anticancer activity in MCF-7 cells and increased oral bioavailability in rats [90].
A systematic review [91] indicated that curcumin reduces the side effects of chemotherapy or radiotherapy, thereby improving the quality of life for patients. Furthermore, the authors reported that curcumin increases patient survival and decreases the level of tumour markers through several molecular pathways including hypoxic stress, angiogenesis, adhesion molecules, and extracellular matrix degradation [91].
Another review highlighted curcumin’s ability to interrupt important stages of tumorigenesis, including proliferation, survival, angiogenesis, and metastasis, in hormone-independent breast cancer, via the modulation of multiple signaling paths. The anticancer activity of curcumin in breast cancer was associated with the PI3K/Akt/mTOR, JAK/STAT, MAPK, NF-ĸB, p53, and Wnt/β-catenin pathways, as well as the apoptosis and cell cycle paths [9].
This systematic review provided a thorough overview of evidence from in vitro and animal model studies on the antitumor effects of curcumin in breast cancer. Our investigation was based on analysis of the five most important databases, with no restrictions imposed on the year of publication and language in the inclusion criteria. We included studies conducted in several countries, including China, India, Turkey, Iran, Italy, USA, Taiwan, Egypt, Bahrain, Romania, and Chile, which helped provide a broad perspective of the topic. However, this review had certain limitations. First, a meta-analysis could not be performed because of the high heterogeneity in the presentation of outcome measures, including different dosing and modes of delivery of curcumin, animal models, and methods of follow-up in the different studies. Furthermore, the adverse effects of curcumin formulations are yet to be investigated thoroughly. As the review did not focus on this aspect, we emphasize the importance of further studies investigating the adverse effects, toxicity, safety, tumor markers, and therapeutic responses in experimental trials and studies conducted on human patients. We believe that the results of ongoing clinical assays will provide a deeper understanding of the therapeutic potential of curcumin as an efficient alternative or adjuvant treatment.

5. Conclusions

This systematic review highlighted the beneficial effects of curcumin against human and animal breast cancer cells with respect to the inhibition of cell proliferation, reduction of malignant cell viability, and induction of apoptosis, and discussed the efficacy of curcumin in tumor growth reduction in experimental breast cancer models. These results were obtained from studies based on the delivery of curcumin via oral administration, implantation, intraperitoneal injection, and nanoparticle formulations. The information presented herein supports randomized clinical trials on the adjuvant properties of curcumin in the treatment of breast mammary neoplasms.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers14092165/s1, Table S1: Search strategies for use in the databases. Table S2: Articles excluded and reasons for exclusion. Table S3: Quality of evidence in the preclinical studies.

Author Contributions

Conceptualization: K.A.B., C.R.M., M.N., A.F.B., C.R.D.F. and M.A.M.S.; Methodology: K.A.B., C.R.M., M.N., A.F.B., C.R.D.F. and M.A.M.S.; Formal Analysis: K.A.B. and C.R.M.; Investigation: K.A.B., C.R.M., M.N., A.F.B., C.R.D.F. and M.A.M.S.; Resources: K.A.B., C.R.M., M.N., A.F.B., C.R.D.F. and M.A.M.S.; Data Curation: K.A.B.; and M.A.M.S.; Writing—Original Draft Preparation: K.A.B., C.R.M. and M.A.M.S.; Writing—Review and Editing: K.A.B., C.R.M., M.N., A.F.B., C.R.D.F. and M.A.M.S.; Visualization: K.A.B., C.R.M., M.N., A.F.B., C.R.D.F. and M.A.M.S.; Supervision: A.F.B. and M.A.M.S.; Project Administration: K.A.B.; Funding Acquisition: M.N., A.F.B. and M.A.M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We thank the Fundação de Amparo à Pesquisa do Estado de Goiás/ Fundação de Apoio à Pesquisa/ Universidade Federal de Goiás (FAPEG/ FUNAPE/UFG), Universidade de Rio Verde (UniRV) and Instituto Federal Goiano (IFG) for partially supporting this study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Momenimovahed, Z.; Salehiniya, H. Epidemiological characteristics of and risk factors for breast cancer in the world. Breast Cancer (Dove Med. Press) 2019, 11, 151–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  3. Li, N.; Wang, Z.; Zhang, Y.; Zhang, K.; Xie, J.; Liu, Y.; Li, W.; Feng, N. Curcumin-loaded redox-responsive mesoporous silica nanoparticles for targeted breast cancer therapy. Artif. Cells Nanomed. Biotechnol. 2018, 46, 921–935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Inotai, A.; Ágh, T.; Maris, R.; Erdősi, D.; Kovács, S.; Kaló, Z.; Senkus, E. Systematic review of real-world studies evaluating the impact of medication non-adherence to endocrine therapies on hard clinical endpoints in patients with non-metastatic breast cancer. Cancer Treat. Rev. 2021, 100, 102264. [Google Scholar] [CrossRef]
  5. Terret, C.; Russo, C. Pharmacotherapeutic Management of Breast Cancer in Elderly Patients: The Promise of Novel Agents. Drugs Aging 2018, 35, 93–115. [Google Scholar] [CrossRef]
  6. Sen, G.S.; Mohanty, S.; Hossain, D.M.S.; Bhattacharyya, S.; Banerjee, S.; Chakraborty, J.; Saha, S.; Ray, P.; Bhattacharjee, P.; Mandal, D.; et al. Curcumin enhances the efficacy of chemotherapy by tailoring p65NFκB-p300 cross-talk in favor of p53–p300 in breast cancer. J. Biol. Chem. 2011, 286, 42232–42247. [Google Scholar] [CrossRef] [Green Version]
  7. Bimonte, S.; Barbieri, A.; Palma, G.; Rea, D.; Luciano, A.; D’Aiuto, M.; Arra, C.; Izzo, F. Dissecting the role of curcumin in tumour growth and angiogenesis in mouse model of human breast cancer. BioMed Res. Int. 2015, 2015, 878134. [Google Scholar] [CrossRef]
  8. Witika, B.A.; Makoni, P.A.; Matafwali, S.K.; Mweetwa, L.L.; Shandele, G.C.; Walker, R.B. Enhancement of Biological and Pharmacological Properties of an Encapsulated Polyphenol: Curcumin. Molecules 2021, 26, 4244. [Google Scholar] [CrossRef]
  9. Farghadani, R.; Naidu, R. Curcumin: Modulator of Key Molecular Signaling Pathways in Hormone-Independent Breast Cancer. Cancers 2021, 13, 3427. [Google Scholar] [CrossRef]
  10. Farghadani, R.; Naidu, R. Curcumin as an Enhancer of Therapeutic Efficiency of Chemotherapy Drugs in Breast Cancer. Int. J. Mol. Sci. 2022, 23, 2144. [Google Scholar] [CrossRef]
  11. Sinha, D.; Biswas, J.; Sung, B.; Aggarwal, B.B.; Bishayee, A. Chemopreventive and chemotherapeutic potential of curcumin in breast cancer. Curr. Drug Targets 2012, 13, 1799–1819. [Google Scholar] [CrossRef] [PubMed]
  12. Song, X.; Zhang, M.; Dai, E.; Luo, Y. Molecular targets of curcumin in breast cancer (Review). Mol. Med. Rep. 2019, 19, 23–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Ombredane, A.S.; Silva, V.R.P.; Andrade, L.R.; Pinheiro, W.O.; Simonelly, M.; Oliveira, J.V.; Pinheiro, A.C.; Gonçalves, G.F.; Felice, G.J.; Garcia, M.P.; et al. In Vivo Efficacy and Toxicity of Curcumin Nanoparticles in Breast Cancer Treatment: A Systematic Review. Front. Oncol. 2021, 11, 612903. [Google Scholar] [CrossRef] [PubMed]
  14. Gianfredi, V.; Nucci, D.; Vannini, S.; Villarini, M.; Moretti, M. In vitro Biological Effects of Sulforaphane (SFN), Epigallocatechin-3-gallate (EGCG), and Curcumin on Breast Cancer Cells: A Systematic Review of the Literature. Nutr. Cancer 2017, 69, 969–978. [Google Scholar] [CrossRef] [PubMed]
  15. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. BMJ 2009, 339, b2535. [Google Scholar] [CrossRef] [Green Version]
  16. de Vries, R.B.M.; Hooijmans, C.R.; Langendam, M.W.; van Luijk, J.; Leenaars, M.; Ritskes-Hoitinga, M.; Wever, K.E. A protocol format for the preparation, registration and publication of systematic reviews of animal intervention studies. Evid. Based Preclin. Med. 2015, 2, e00007. [Google Scholar] [CrossRef]
  17. Yang, N.; Ray, S.D.; Krafts, K. Cell Proliferation. In Encyclopedia of Toxicology, 3rd ed.; Wexler, P., Ed.; Academic Press: Oxford, UK, 2014. [Google Scholar]
  18. Peng, Y.; Li, J.; Zhu, L. Chapter 8—Cancer and non-coding RNAs. In Nutritional Epigenomics; Ferguson, B.S., Ed.; Academic Press: Oxford, UK, 2019; Volume 14, pp. 119–132. [Google Scholar]
  19. Fang, I.J.; Trewyn, B.G. Application of mesoporous silica nanoparticles in intracellular delivery of molecules and proteins. Methods Enzymol. 2012, 508, 41–59. [Google Scholar]
  20. Khan, Y. Characterizing the Properties of Tissue Constructs for Regenerative Engineering; Elsevier: Amsterdam, The Netherlands, 2019. [Google Scholar] [CrossRef]
  21. Brown, D.A.; Yang, N.; Ray, S.D. Apoptosis. In Encyclopedia of Toxicology, 3rd ed.; Wexler, P., Ed.; Academic Press: Oxford, UK, 2014. [Google Scholar]
  22. Wong, R.S. Apoptosis in cancer: From pathogenesis to treatment. J. Exp. Clin. Cancer Res. 2011, 30, 87. [Google Scholar] [CrossRef] [Green Version]
  23. Noll, M.; Wedderkopp, N.; Mendonça, C.R.; Kjaer, P. Motor performance and back pain in children and adolescents: A systematic review and meta-analysis protocol. Syst. Rev. 2020, 9, 212. [Google Scholar] [CrossRef]
  24. Noll, M.; Kjaer, P.; Mendonça, C.R.; Wedderkopp, N. Motor performance and back pain in children and adolescents: A systematic review. Eur. J. Pain 2022, 26, 77–102. [Google Scholar] [CrossRef]
  25. Pavan, L.M.C.; Rêgo, D.F.; Elias, S.T.; De Luca Canto, G.; Guerra, E.N.S. In vitro Anti-Tumor Effects of Statins on Head and Neck Squamous Cell Carcinoma: A Systematic Review. PLoS ONE 2015, 10, e0130476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Wei, D.; Tang, K.; Wang, Q.; Estill, J.; Yao, L.; Wang, X.; Chen, Y.; Yang, K. The use of GRADE approach in systematic reviews of animal studies. J. Evid.-Based Med. 2016, 9, 98–104. [Google Scholar] [CrossRef] [PubMed]
  27. Abbaspour, H.; Safipour Afshar, A. Curcumin inhibits the expression of ornithine decarboxylase and adenosine deaminase genes in MCF-7 human breast cancer cells. Arch. Biol. Sci. 2018, 70, 639–645. [Google Scholar] [CrossRef]
  28. Abuelba, H.; Cotrutz, C.E.; Stoica, B.A.; Stoica, L.; Olinici, D.; Petreuş, T. In vitro evaluation of curcumin effects on breast adenocarcinoma 2D and 3D cell cultures. Rom. J. Morphol. Embryol. = Rev. Roum. De Morphol. Et Embryol. 2015, 56, 71–76. [Google Scholar]
  29. Alizadeh, A.M.; Sadeghizadeh, M.; Najafi, F.; Ardestani, S.K.; Erfani-Moghadam, V.; Khaniki, M.; Rezaei, A.; Zamani, M.; Khodayari, S.; Khodayari, H.; et al. Encapsulation of Curcumin in Diblock Copolymer Micelles for Cancer Therapy. BioMed Res. Int. 2015, 2015, 824746. [Google Scholar] [CrossRef] [PubMed]
  30. Bansal, S.S.; Kausar, H.; Vadhanam, M.V.; Ravoori, S.; Pan, J.; Rai, S.N.; Gupta, R.C. Curcumin implants, not curcumin diet, inhibit estrogen-induced mammary carcinogenesis in ACI rats. Cancer Prev. Res. 2014, 7, 456–465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Calaf, G.M.; Ponce-Cusi, R.; Carrión, F. Curcumin and paclitaxel induce cell death in breast cancer cell lines. Oncol. Rep. 2018, 40, 2381–2388. [Google Scholar] [CrossRef]
  32. Chen, W.; Li, L.; Zhang, X.; Liang, Y.-k.; Pu, Z.; Wang, L.; Mo, J.J.D.D. Curcumin: A calixarene derivative micelle potentiates anti-breast cancer stem cells effects in xenografted, triple-negative breast cancer mouse models. Drug Deliv. 2017, 24, 1470–1481. [Google Scholar] [CrossRef] [Green Version]
  33. Chiu, T.L.; Su, C.C. Curcumin inhibits proliferation and migration by increasing the Bax to Bcl-2 ratio and decreasing NF-kappaBp65 expression in breast cancer MDA-MB-231 cells. Int. J. Mol. Med. 2009, 23, 469–475. [Google Scholar]
  34. Choudhuri, T.; Pal, S.; Agwarwal, M.L.; Das, T.; Sa, G. Curcumin induces apoptosis in human breast cancer cells through p53-dependent Bax induction. FEBS Lett. 2002, 512, 334–340. [Google Scholar] [CrossRef] [Green Version]
  35. Coker-Gurkan, A.; Bulut, D.; Genc, R.; Arisan, E.D.; Obakan-Yerlikaya, P.; Palavan-Unsal, N. Curcumin prevented human autocrine growth hormone (GH) signaling mediated NF-κB activation and miR-183-96-182 cluster stimulated epithelial mesenchymal transition in T47D breast cancer cells. Mol. Biol. Rep. 2019, 46, 355–369. [Google Scholar] [CrossRef] [PubMed]
  36. Coker-Gurkan, A.; Celik, M.; Ugur, M.; Arisan, E.D.; Obakan-Yerlikaya, P.; Durdu, Z.B.; Palavan-Unsal, N. Curcumin inhibits autocrine growth hormone-mediated invasion and metastasis by targeting NF-κB signaling and polyamine metabolism in breast cancer cells. Amino Acids 2018, 50, 1045–1069. [Google Scholar] [CrossRef] [PubMed]
  37. Fan, H.; Liang, Y.; Jiang, B.; Li, X.; Xun, H.; Sun, J.; He, W.; Lau, H.T.; Ma, X. Curcumin inhibits intracellular fatty acid synthase and induces apoptosis in human breast cancer MDA-MB-231 cells. Oncol. Rep. 2016, 35, 2651–2656. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Fathy Abd-Ellatef, G.E.; Gazzano, E.; Chirio, D.; Hamed, A.R.; Belisario, D.C.; Zuddas, C.; Peira, E.; Rolando, B.; Kopecka, J.; Assem Said Marie, M.; et al. Curcumin-Loaded Solid Lipid Nanoparticles Bypass P-Glycoprotein Mediated Doxorubicin Resistance in Triple Negative Breast Cancer Cells. Pharmaceutics 2020, 12, 96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Ghosh, S.; Dutta, S.; Sarkar, A.; Kundu, M.; Sil, P.C. Targeted delivery of curcumin in breast cancer cells via hyaluronic acid modified mesoporous silica nanoparticle to enhance anticancer efficiency. Colloids Surf. B Biointerfaces 2021, 197, 111404. [Google Scholar] [CrossRef] [PubMed]
  40. Greish, K.; Pittalà, V.; Taurin, S.; Taha, S.; Bahman, F.; Mathur, A.; Jasim, A.; Mohammed, F.; El-Deeb, I.M.; Fredericks, S.; et al. Curcumin-Copper Complex Nanoparticles for the Management of Triple-Negative Breast Cancer. Nanomaterials 2018, 8, 884. [Google Scholar] [CrossRef] [Green Version]
  41. Grill, A.E.; Shahani, K.; Koniar, B.; Panyam, J. Chemopreventive efficacy of curcumin-loaded PLGA microparticles in a transgenic mouse model of HER-2-positive breast cancer. Drug Deliv. Transl. Res. 2018, 8, 329–341. [Google Scholar] [CrossRef] [PubMed]
  42. Hashemzehi, M.; Behnam-Rassouli, R.; Hassanian, S.M.; Moradi-Binabaj, M.; Moradi-Marjaneh, R.; Rahmani, F.; Fiuji, H.; Jamili, M.; Mirahmadi, M.; Boromand, N.; et al. Phytosomal-curcumin antagonizes cell growth and migration, induced by thrombin through AMP-Kinase in breast cancer. J. Cell Biochem. 2018, 119, 5996–6007. [Google Scholar] [CrossRef]
  43. He, H.; Zhuang, W.; Ma, B.; Su, X.; Yu, T.; Hu, J.; Chen, L.; Peng, R.; Li, G.; Wang, Y. Oxidation-Responsive and Aggregation-Induced Emission Polymeric Micelles with Two-Photon Excitation for Cancer Therapy and Bioimaging. ACS Biomater. Sci. Eng. 2019, 5, 2577–2586. [Google Scholar] [CrossRef]
  44. Hu, S.; Xu, Y.; Meng, L.; Huang, L.; Sun, H. Curcumin inhibits proliferation and promotes apoptosis of breast cancer cells. Exp. Ther. Med. 2018, 16, 1266–1272. [Google Scholar] [CrossRef]
  45. Hua, W.F.; Fu, Y.S.; Liao, Y.J.; Xia, W.J.; Chen, Y.C.; Zeng, Y.X.; Kung, H.F.; Xie, D. Curcumin induces down-regulation of EZH2 expression through the MAPK pathway in MDA-MB-435 human breast cancer cells. Eur. J. Pharmacol. 2010, 637, 16–21. [Google Scholar] [CrossRef] [PubMed]
  46. Huang, C.; Chen, F.; Zhang, L.; Yang, Y.; Yang, X.; Pan, W. (99 m)Tc Radiolabeled HA/TPGS-Based Curcumin-Loaded Nanoparticle for Breast Cancer Synergistic Theranostics: Design, in vitro and in vivo Evaluation. Int. J. Nanomed. 2020, 15, 2987–2998. [Google Scholar] [CrossRef] [PubMed]
  47. Ji, P.; Wang, L.; Chen, Y.; Wang, S.; Wu, Z.; Qi, X. Hyaluronic acid hydrophilic surface rehabilitating curcumin nanocrystals for targeted breast cancer treatment with prolonged biodistribution. Biomater. Sci. 2020, 8, 462–472. [Google Scholar] [CrossRef] [PubMed]
  48. Jiang, M.; Huang, O.; Zhang, X.; Xie, Z.; Shen, A.; Liu, H.; Geng, M.; Shen, K. Curcumin induces cell death and restores tamoxifen sensitivity in the antiestrogen-resistant breast cancer cell lines MCF-7/LCC2 and MCF-7/LCC9. Molecules 2013, 18, 701–720. [Google Scholar] [CrossRef] [Green Version]
  49. Jin, H.; Pi, J.; Zhao, Y.; Jiang, J.; Li, T.; Zeng, X.; Yang, P.; Evans, C.E.; Cai, J. EGFR-targeting PLGA-PEG nanoparticles as a curcumin delivery system for breast cancer therapy. Nanoscale 2017, 9, 16365–16374. [Google Scholar] [CrossRef]
  50. Jung, K.H.; Lee, J.H.; Park, J.W.; Kim, D.H.; Moon, S.H.; Cho, Y.S.; Lee, K.H. Targeted therapy of triple negative MDA-MB-468 breast cancer with curcumin delivered by epidermal growth factor-conjugated phospholipid nanoparticles. Oncol. Lett. 2018, 15, 9093–9100. [Google Scholar] [CrossRef] [Green Version]
  51. Kim, J.M.; Noh, E.M.; Kwon, K.B.; Kim, J.S.; You, Y.O.; Hwang, J.K.; Hwang, B.M.; Kim, B.S.; Lee, S.H.; Lee, S.J.; et al. Curcumin suppresses the TPA-induced invasion through inhibition of PKCα-dependent MMP-expression in MCF-7 human breast cancer cells. Phytomedicine 2012, 19, 1085–1092. [Google Scholar] [CrossRef]
  52. Kumari, P.; Muddineti, O.S.; Rompicharla, S.V.; Ghanta, P.; Karthik, B.B.N.A.; Ghosh, B.; Biswas, S. Cholesterol-conjugated poly (D, L-lactide)-based micelles as a nanocarrier system for effective delivery of curcumin in cancer therapy. Drug Deliv. 2017, 24, 209–223. [Google Scholar] [CrossRef] [Green Version]
  53. Kumari, P.; Paul, M.; Bobde, Y.; Soniya, K.; Kiran Rompicharla, S.V.; Ghosh, B.; Biswas, S. Albumin-based lipoprotein nanoparticles for improved delivery and anticancer activity of curcumin for cancer treatment. Nanomedicine 2020, 15, 2851–2869. [Google Scholar] [CrossRef]
  54. Kumari, P.; Swami, M.O.; Nadipalli, S.K.; Myneni, S.; Ghosh, B.; Biswas, S. Curcumin Delivery by Poly(Lactide)-Based Co-Polymeric Micelles: An In Vitro Anticancer Study. Pharm. Res. 2016, 33, 826–841. [Google Scholar] [CrossRef]
  55. Laha, D.; Pal, K.; Ray Chowdhuri, A.; Parida, P.; Sahu, S.; Jana, K.; Karmakar, P. Fabrication of curcumin loaded folic acid tagged metal organic framework for triple negative breast cancer therapy in in vitro and in vivo system. New J. Chem. 2018, 43, 217–229. [Google Scholar] [CrossRef]
  56. Lai, H.-W.; Chien, S.-Y.; Kuo, S.-J.; Tseng, L.-M.; Lin, H.-Y.; Chi, C.-W.; Chen, D.-R. The Potential Utility of Curcumin in the Treatment of HER-2-Overexpressed Breast Cancer: An In Vitro and In Vivo Comparison Study with Herceptin. Evid. Based Complement. Altern. Med. 2012, 2012, 486568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Lin, M.; Teng, L.; Wang, Y.; Zhang, J.; Sun, X. Curcumin-guided nanotherapy: A lipid-based nanomedicine for targeted drug delivery in breast cancer therapy. Drug Deliv. 2016, 23, 1420–1425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Liu, L.; Sun, L.; Wu, Q.; Guo, W.; Li, L.; Chen, Y.; Li, Y.; Gong, C.; Qian, Z.; Wei, Y. Curcumin loaded polymeric micelles inhibit breast tumor growth and spontaneous pulmonary metastasis. Int. J. Pharm. 2013, 443, 175–182. [Google Scholar] [CrossRef] [PubMed]
  59. Liu, Q.; Loo, W.T.; Sze, S.C.; Tong, Y. Curcumin inhibits cell proliferation of MDA-MB-231 and BT-483 breast cancer cells mediated by down-regulation of NFkappaB, cyclinD and MMP-1 transcription. Phytomedicine 2009, 16, 916–922. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Lv, L.; Guo, Y.; Shen, Y.; Liu, J.; Zhang, W.; Zhou, D.; Guo, S. Intracellularly Degradable, Self-Assembled Amphiphilic Block Copolycurcumin Nanoparticles for Efficient In Vivo Cancer Chemotherapy. Adv. Healthc. Mater. 2015, 4, 1496–1501. [Google Scholar] [CrossRef] [Green Version]
  61. Lv, Z.D.; Liu, X.P.; Zhao, W.J.; Dong, Q.; Li, F.N.; Wang, H.B.; Kong, B. Curcumin induces apoptosis in breast cancer cells and inhibits tumor growth in vitro and in vivo. Int. J. Clin. Exp. Pathol. 2014, 7, 2818–2824. [Google Scholar]
  62. Mahalunkar, S.; Yadav, A.S.; Gorain, M.; Pawar, V.; Braathen, R.; Weiss, S.; Bogen, B.; Gosavi, S.W.; Kundu, G.C. Functional design of pH-responsive folate-targeted polymer-coated gold nanoparticles for drug delivery and in vivo therapy in breast cancer. Int. J. Nanomed. 2019, 14, 8285–8302. [Google Scholar] [CrossRef] [Green Version]
  63. Masuelli, L.; Benvenuto, M.; Fantini, M.; Marzocchella, L.; Sacchetti, P.; Di Stefano, E.; Tresoldi, I.; Izzi, V.; Bernardini, R.; Palumbo, C.; et al. Curcumin induces apoptosis in breast cancer cell lines and delays the growth of mammary tumors in neu transgenic mice. J. Biol. Regul. Homeost. Agents 2013, 27, 105–119. [Google Scholar]
  64. Mehta, K.; Pantazis, P.; McQueen, T.; Aggarwal, B.B. Antiproliferative effect of curcumin (diferuloylmethane) against human breast tumor cell lines. Anti-Cancer Drugs 1997, 8, 470–481. [Google Scholar] [CrossRef]
  65. Montazeri, M.; Pilehvar-Soltanahmadi, Y.; Mohaghegh, M.; Panahi, A.; Khodi, S.; Zarghami, N.; Sadeghizadeh, M. Antiproliferative and Apoptotic Effect of Dendrosomal Curcumin Nanoformulation in P53 Mutant and Wide-Type Cancer Cell Lines. Anticancer Agents Med. Chem. 2017, 17, 662–673. [Google Scholar] [CrossRef]
  66. Mukerjee, A.; Ranjan, A.P.; Vishwanatha, J.K. Targeted Nanocurcumin Therapy Using Annexin A2 Anitbody Improves Tumor Accumulation and Therapeutic Efficacy Against Highly Metastatic Breast Cancer. J. Biomed. Nanotechnol. 2016, 12, 1374–1392. [Google Scholar] [CrossRef]
  67. Mukhopadhyay, R.; Sen, R.; Paul, B.; Kazi, J.; Ganguly, S.; Debnath, M.C. Gemcitabine Co-Encapsulated with Curcumin in Folate Decorated PLGA Nanoparticles; a Novel Approach to Treat Breast Adenocarcinoma. Pharm. Res. 2020, 37, 56. [Google Scholar] [CrossRef]
  68. Pal, K.; Laha, D.; Parida, P.K.; Roy, S.; Bardhan, S.; Dutta, A.; Jana, K.; Karmakar, P. An In Vivo Study for Targeted Delivery of Curcumin in Human Triple Negative Breast Carcinoma Cells Using Biocompatible PLGA Microspheres Conjugated with Folic Acid. J. Nanosci. Nanotechnol. 2019, 19, 3720–3733. [Google Scholar] [CrossRef]
  69. Sahne, F.; Mohammadi, M.; Najafpour, G.D. Single-Layer Assembly of Multifunctional Carboxymethylcellulose on Graphene Oxide Nanoparticles for Improving in Vivo Curcumin Delivery into Tumor Cells. ACS Biomater. Sci. Eng. 2019, 5, 2595–2609. [Google Scholar] [CrossRef]
  70. Sarighieh, M.A.; Montazeri, V.; Shadboorestan, A.; Ghahremani, M.H.; Ostad, S.N. The Inhibitory Effect of Curcumin on Hypoxia Inducer Factors (Hifs) as a Regulatory Factor in the Growth of Tumor Cells in Breast Cancer Stem-Like Cells. Drug Res. 2020, 70, 512–518. [Google Scholar] [CrossRef]
  71. Shiri, S.; Alizadeh, A.M.; Baradaran, B.; Farhanghi, B.; Shanehbandi, D.; Khodayari, S.; Khodayari, H.; Tavassoli, A. Dendrosomal curcumin suppresses metastatic breast cancer in mice by changing m1/m2 macrophage balance in the tumor microenvironment. Asian Pac. J. Cancer Prev. 2015, 16, 3917–3922. [Google Scholar] [CrossRef] [Green Version]
  72. Shukla, M.; Jaiswal, S.; Sharma, A.; Srivastava, P.K.; Arya, A.; Dwivedi, A.K.; Lal, J. A combination of complexation and self-nanoemulsifying drug delivery system for enhancing oral bioavailability and anticancer efficacy of curcumin. Drug Dev. Ind. Pharm. 2017, 43, 847–861. [Google Scholar] [CrossRef]
  73. Sun, S.H.; Huang, H.C.; Huang, C.; Lin, J.K. Cycle arrest and apoptosis in MDA-MB-231/Her2 cells induced by curcumin. Eur. J. Pharmacol. 2012, 690, 22–30. [Google Scholar] [CrossRef]
  74. Sun, X.D.; Liu, X.E.; Huang, D.S. Curcumin induces apoptosis of triple-negative breast cancer cells by inhibition of EGFR expression. Mol. Med. Rep. 2012, 6, 1267–1270. [Google Scholar] [CrossRef] [Green Version]
  75. Vakilinezhad, M.A.; Amini, A.; Dara, T.; Alipour, S. Methotrexate and Curcumin co-encapsulated PLGA nanoparticles as a potential breast cancer therapeutic system: In vitro and in vivo evaluation. Colloids Surfaces. B Biointerfaces 2019, 184, 110515. [Google Scholar] [CrossRef] [PubMed]
  76. Wang, X.; Hang, Y.; Liu, J.; Hou, Y.; Wang, N.; Wang, M. Anticancer effect of curcumin inhibits cell growth through miR-21/PTEN/Akt pathway in breast cancer cell. Oncol. Lett. 2017, 13, 4825–4831. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Wang, Y.; Luo, Z.; Wang, Z.; You, M.; Xie, S.; Peng, Y.; Yang, H. Effect of curcumin-loaded nanoparticles on mitochondrial dysfunctions of breast cancer cells. J. Nanopart. Res. 2018, 20, 1–11. [Google Scholar] [CrossRef]
  78. Yang, Z.; Sun, N.; Cheng, R.; Zhao, C.; Liu, J.; Tian, Z. Hybrid nanoparticles coated with hyaluronic acid lipoid for targeted co-delivery of paclitaxel and curcumin to synergistically eliminate breast cancer stem cells. J. Mater. Chem. B 2017, 5, 6762–6775. [Google Scholar] [CrossRef]
  79. Yang, Z.; Sun, N.; Cheng, R.; Zhao, C.; Liu, Z.; Li, X.; Liu, J.; Tian, Z. pH multistage responsive micellar system with charge-switch and PEG layer detachment for co-delivery of paclitaxel and curcumin to synergistically eliminate breast cancer stem cells. Biomaterials 2017, 147, 53–67. [Google Scholar] [CrossRef]
  80. Younesian, O.; Kazerouni, F.; Dehghan-Nayeri, N.; Omrani, D.; Rahimipour, A.; Shanaki, M.; Rezapour Kalkhoran, M.; Cheshmi, F. Effect of Curcumin on Fatty Acid Synthase Expression and Enzyme Activity in Breast Cancer Cell Line SKBR3. Int. J. Cancer Manag. 2017, 10, e8173. [Google Scholar] [CrossRef]
  81. Yu, S.; Wang, S.; Xie, Z.; Yu, S.; Li, L.; Xiao, H.; Song, Y. Hyaluronic acid coating on the surface of curcumin-loaded ZIF-8 nanoparticles for improved breast cancer therapy: An in vitro and in vivo study. Colloids Surf. B Biointerfaces 2021, 203, 111759. [Google Scholar] [CrossRef]
  82. Yu, Y.; Zhang, X.; Qiu, L. The anti-tumor efficacy of curcumin when delivered by size/charge-changing multistage polymeric micelles based on amphiphilic poly(β-amino ester) derivates. Biomaterials 2014, 35, 3467–3479. [Google Scholar] [CrossRef]
  83. Yuan, J.D.; ZhuGe, D.L.; Tong, M.Q.; Lin, M.T.; Xu, X.F.; Tang, X.; Zhao, Y.Z.; Xu, H.L. pH-sensitive polymeric nanoparticles of mPEG-PLGA-PGlu with hybrid core for simultaneous encapsulation of curcumin and doxorubicin to kill the heterogeneous tumour cells in breast cancer. Artif. Cells Nanomed. 2018, 46, 302–313. [Google Scholar] [CrossRef] [Green Version]
  84. Zong, H.; Wang, F.; Fan, Q.X.; Wang, L.X. Curcumin inhibits metastatic progression of breast cancer cell through suppression of urokinase-type plasminogen activator by NF-kappa B signaling pathways. Mol. Biol. Rep. 2012, 39, 4803–4808. [Google Scholar] [CrossRef]
  85. Liu, H.-T.; Ho, Y.-S. Anticancer effect of curcumin on breast cancer and stem cells. Food Sci. Hum. Wellness 2018, 7, 134–137. [Google Scholar] [CrossRef]
  86. Abbasalizadeh, F.; Alizadeh, E.; Bagher Fazljou, S.M.; Torbati, M.; Akbarzadeh, A. Anticancer Effect of Alginate-Chitosan Hydrogel Loaded with Curcumin and Chrysin on Lung and Breast Cancer Cell Lines. Curr. Drug Deliv. 2021. [Google Scholar] [CrossRef]
  87. Saghatelyan, T.; Tananyan, A.; Janoyan, N.; Tadevosyan, A.; Petrosyan, H.; Hovhannisyan, A.; Hayrapetyan, L.; Arustamyan, M.; Arnhold, J.; Rotmann, A.R.; et al. Efficacy and safety of curcumin in combination with paclitaxel in patients with advanced, metastatic breast cancer: A comparative, randomized, double-blind, placebo-controlled clinical trial. Phytomedicine 2020, 70, 153218. [Google Scholar] [CrossRef]
  88. Stohs, S.J.; Ji, J.; Bucci, L.R.; Preuss, H.G. A Comparative Pharmacokinetic Assessment of a Novel Highly Bioavailable Curcumin Formulation with 95% Curcumin: A Randomized, Double-Blind, Crossover Study. J. Am. Coll. Nutr. 2018, 37, 51–598. [Google Scholar] [CrossRef] [Green Version]
  89. Stohs, S.J.; Chen, O.; Ray, S.D.; Ji, J.; Bucci, L.R.; Preuss, H.G. Highly Bioavailable Forms of Curcumin and Promising Avenues for Curcumin-Based Research and Application: A Review. Molecules 2020, 25, 1397. [Google Scholar] [CrossRef] [Green Version]
  90. Giordano, A.; Tommonaro, G. Curcumin and Cancer. Nutrients 2019, 5, 2376. [Google Scholar] [CrossRef] [Green Version]
  91. Mansouri, K.; Rasoulpoor, S.; Daneshkhah, A.; Abolfathi, S.; Salari, N.; Mohammadi, M.; Rasoulpoor, S.; Shabani, S. Clinical effects of curcumin in enhancing cancer therapy: A systematic review. BMC Cancer 2020, 24, 791. [Google Scholar] [CrossRef]
Figure 1. Flowchart for study selection (PRISMA Flow Diagram 2020).
Figure 1. Flowchart for study selection (PRISMA Flow Diagram 2020).
Cancers 14 02165 g001
Figure 2. Cellular and molecular mechanisms of action of curcumin in breast cancer. Curcumin exerts its anticancer effect by modulating cell proliferation, inducing apoptosis and inhibiting cancer spread. JAK: janus kinase, STAT: signal transducer and activator of transcription, IL-6: interleukin-6, IκKB: inhibitor of kappa B kinase, TGF: transforming growth factor, EGFR: epidermal growth factor receptor, MAPK: mitogen-activated protein kinase, MAPKK: MAPK kinase, JNK: c-Jun N-terminal kinases, Bcl-2: B-cell lymphoma 2, Bak: Bcl-2 homologous antagonist/killer, Bad: BCL2 associated agonist of cell death, Bid: BH3 interacting-domain death agonist, Bax: Bcl-2 associated X protein, Bcl-xL: ROS: reactive oxygen species, NF-κB: nuclear factor-κ-gene binding, COX-2: Cyclooxygenase 2, ERK1/2: extracellular regulated protein kinase 1 and 2, PI3K: phosphatidylinositol 3-kinase, Akt: protein kinase B, mTOR: mammalian target of rapamycin, JNK: Jun N-terminal kinase, FADD: Fas-associated protein with death domain, p38: mitogen-activated protein kinases, FAZ/CD95: type-II transmembrane protein that belongs to the tumor necrosis fator, Caspases: cysteine-dependent aspartate-specific protease, p53: tumor-suppressor protein, ↓: inhibition, ↑: activation.
Figure 2. Cellular and molecular mechanisms of action of curcumin in breast cancer. Curcumin exerts its anticancer effect by modulating cell proliferation, inducing apoptosis and inhibiting cancer spread. JAK: janus kinase, STAT: signal transducer and activator of transcription, IL-6: interleukin-6, IκKB: inhibitor of kappa B kinase, TGF: transforming growth factor, EGFR: epidermal growth factor receptor, MAPK: mitogen-activated protein kinase, MAPKK: MAPK kinase, JNK: c-Jun N-terminal kinases, Bcl-2: B-cell lymphoma 2, Bak: Bcl-2 homologous antagonist/killer, Bad: BCL2 associated agonist of cell death, Bid: BH3 interacting-domain death agonist, Bax: Bcl-2 associated X protein, Bcl-xL: ROS: reactive oxygen species, NF-κB: nuclear factor-κ-gene binding, COX-2: Cyclooxygenase 2, ERK1/2: extracellular regulated protein kinase 1 and 2, PI3K: phosphatidylinositol 3-kinase, Akt: protein kinase B, mTOR: mammalian target of rapamycin, JNK: Jun N-terminal kinase, FADD: Fas-associated protein with death domain, p38: mitogen-activated protein kinases, FAZ/CD95: type-II transmembrane protein that belongs to the tumor necrosis fator, Caspases: cysteine-dependent aspartate-specific protease, p53: tumor-suppressor protein, ↓: inhibition, ↑: activation.
Cancers 14 02165 g002
Table 3. Risk of bias according to the SYRCLE’s RoB Toll criteria for animal models.
Table 3. Risk of bias according to the SYRCLE’s RoB Toll criteria for animal models.
AuthorsSelection BiasPerformance BiasDetection BiasAttrition BiasReporting BiasOther Biases
12345678910
Abd-Ellatef et al., 2020 [38] Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Alizadeh et al., 2015 [29] Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Bansal et al., 2014 [30] Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Bimonte et al., 2015 [7] Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i002 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Chen et al., 2017 [32] Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Ghosh et al., 2021 [39] Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Greish et al., 2018 [40] Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Grill et al., 2018 [41] Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Hashemzehi et al., 2018 [42] Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
He et al., 2019 [43] Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i001 Cancers 14 02165 i003
Huang et al., 2020 [46] China Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Ji et al., 2020 [47] Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Jin et al., 2017 [49] Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Jung et al., 2018 [50] Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i002 Cancers 14 02165 i001 Cancers 14 02165 i003
Kumari et al., 2020 [52] Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Laha et al., 2018 [55] Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Lai et al., 2012 [56] Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Li et al., 2018 [3] Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Lin et al., 2016 [57] Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Liu et al., 2013 [58] Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Lv et al., 2014 [61] Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003
Lv et al., 2015 [60] Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i002 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003
Mahalunkar et al., 2019 [62] Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Masuelli et al., 2013 [63] Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Mukerjee et al., 2016 [66] USA Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Mukhopadhyay et al., 2020 [67] Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Pal et al., 2019 [68] Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Sahne et al., 2019 [69] Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Shiri et al., 2015 [71] Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Shukla et al., 2017 [72] Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i002 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Vakilinezhad et al., 2019 [75] Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Wang et al., 2018 [77] Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Yang et al., 2017 a [78] Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Yang et al., 2017 b [79] Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003
Yu et al., 2014 [82] Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Yu et al., 2021 [81] Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
Yuan et al., 2018 [83] Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i003 Cancers 14 02165 i001 Cancers 14 02165 i003
YES Cancers 14 02165 i001 NO Cancers 14 02165 i002 UNCLEAR Cancers 14 02165 i003. YES indicates low risk of bias; NO indicates high risk of bias; UNCLEAR indicates inability of bias assignment. The ten items assessed included: 1 Was the sequence of assignment generated and applied properly? 2 Were the groups similar at baseline, or were they adjusted for confounders in the analysis? 3 Was the allocation to the different groups adequately concealed? 4 Were the animals randomly housed during the experiment? 5 Were caregivers and/or investigators blinded to the intervention each animal received during the experiment? 6 Were the animals randomly selected for the evaluation of results? 7 Was the outcome assessor blinded? 8 Were data of incomplete results handled appropriately? 9 Are study reports exempt from selective result reporting? 10 Was the study apparently free from other problems that could cause a high risk of bias?
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Barcelos, K.A.; Mendonça, C.R.; Noll, M.; Botelho, A.F.; Francischini, C.R.D.; Silva, M.A.M. Antitumor Properties of Curcumin in Breast Cancer Based on Preclinical Studies: A Systematic Review. Cancers 2022, 14, 2165. https://doi.org/10.3390/cancers14092165

AMA Style

Barcelos KA, Mendonça CR, Noll M, Botelho AF, Francischini CRD, Silva MAM. Antitumor Properties of Curcumin in Breast Cancer Based on Preclinical Studies: A Systematic Review. Cancers. 2022; 14(9):2165. https://doi.org/10.3390/cancers14092165

Chicago/Turabian Style

Barcelos, Kênia Alves, Carolina Rodrigues Mendonça, Matias Noll, Ana Flávia Botelho, Cristiane Raquel Dias Francischini, and Marco Augusto Machado Silva. 2022. "Antitumor Properties of Curcumin in Breast Cancer Based on Preclinical Studies: A Systematic Review" Cancers 14, no. 9: 2165. https://doi.org/10.3390/cancers14092165

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop